PROGNOSTIC TUMOR BIOMARKERS

Prognostic and predictive biomarkers are disclosed that can be used in systems and methods for predicting the prognosis of a subject with a cancer and to direct therapy based on that prognosis.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims benefit of U.S. Provisional Application No. 62/055,415, filed Sep. 25, 2014, and U.S. Provisional Application Ser. No. 62/083,586, filed Nov. 24, 2014, which are hereby incorporated herein by reference in their entirety.

BACKGROUND

Cancer patients and their loved ones face many unknowns. Understanding their disease and what to expect can help patients and their loved ones make decisions about treatment, supportive and palliative care, rehabilitation, and personal matters, such as financial matters.

Many factors can influence the prognosis of a person with cancer. Among the most important are the type and location of the cancer, the stage of the disease (the extent to which the cancer has spread in the body), and the cancer's grade (how abnormal the cancer cells look under a microscope—an indicator of how quickly the cancer is likely to grow and spread). Other factors that affect prognosis include the biological and genetic properties of the cancer cells, the patient's age and overall general health, and the extent to which the patient's cancer responds to treatment.

Improved biomarkers and methods are needed to provide accurate and personalized prognosis for cancer patients.

SUMMARY

Prognostic and predictive biomarkers are disclosed that were identified from gene expression profiling data from approximately 16,000 cancer subjects. These data were split into two parts. The first part, in combination with patient clinical data, was used to discover prognostic and predictive biomarkers for a series of different cancers capable and to train risk prediction models. These models were then validated using the second part of the gene expression profiling data. Therefore, systems and methods of using these biomarkers and predictive models are disclosed.

For example, a method for predicting prognosis of a patient with breast cancer is disclosed that involves the use of a composite model to predict the risk of bone metastasis and death. The method involves first determining gene expression intensities for several signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is estrogen receptor (ER) gene expression. In some embodiments, one of the components is human epidermal growth factor receptor 2 (HER2) gene expression. In some embodiments, one of the components is a proliferation signature gene score. This proliferation signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 1, or genes highly correlated to the mean log expression of genes in Table 1, such as TPX2, CENPA, KIF2C, CCNB2, BUB1, HJURP, CDCA5, PTTG1, CEP55, and SKA1. In some embodiments, one of the components is an immune signature gene score. This immune signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 2, or genes highly correlated to the mean log expression of genes in Table 2, such as CD3D, CD2, CD3E, ITK, TRBC1, TBC1D10C, ACAP1, CD247, SLAMF6, and IKZF1. The method can then involve calculating a breast cancer risk score from the gene expression intensities of each category, e.g., such that a high breast cancer risk score is an indication that the subject has a high risk for bone metastasis and/or death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. A more aggressive treatment for high score patients may include chemotherapy and bone metastasis preventive therapies like bisphosphonates, antibodies to RANKL or DKK1. For ER+ patients, more aggressive treatment for high score patients may include mTOR inhibitors, immune therapy like PD-1 inhibitors. For ER—patients, immune signature predicts relatively good outcome, so low-risk score in ER—maybe a selection factor for immune therapies like PD-1 or CTLA4 inhibitors. High risk patients could also be preferentially considered for genetic tests for targeted therapies like inhibitors for PI3K/AKT pathway. Patients with high immune signatures could be selected for immune therapies like anti-PD1. This prognostic model can be used to identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with lung cancer that also involves the use of a composite model to predict the risk of death. This method also involves first determining gene expression intensities for several signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is an immune signature gene score. This immune signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 4, or genes highly correlated to the mean log expression of genes in Table 4, such as, CD2, ITGAL, IKZF1, CD3D, TRBC1, ACAP1, CD3E, TBC1D10C, CD247, and SLAMF6. In some embodiments, one of the components is a hypoxia signature gene score. This hypoxia signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 5, or genes highly correlated to the mean log expression of genes in Table 5, such as SLC2A1, S100A2, KRT16, KRT6A, CD109, GJB3, SFN, MICALL1, RNTL2, and COL7A1. In some embodiments, one of the components is a lung cancer prognosis signature gene score. This lung cancer prognosis signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 7, or genes highly correlated to the mean log expression of genes in Table 7, such as HLF, SCN7A, NR3C2, PCDP1, ABCA8, EMCN, IFT57, BDH2, MAMDC2, and ITGA8. In some embodiments, one of the components is a proliferation signature gene score. This proliferation score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 8, or genes highly correlated to the mean log expression of genes in Table 8, such as TPX2, CENPA, KIF2C, CCNB2, CDCA5, HJURP, KIF4A, BIRC5, DLGAP5, and SKA1. The method can further involve determining the composite tumor stage. The method can then involve calculating a lung cancer risk score from the gene expression intensities of each category and the composite tumor stage, e.g., such that a high lung cancer risk score is an indication that the subject has a high risk for death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. For example, patients with high risk scores can be more aggressively treated with chemotherapies like cisplatin, carboplatin, docetaxel, or combinations. These patients could also be preferentially considered for genetic tests for targeted therapies like EGFR inhibitors or ALK inhibitors. Patients with high immune signatures could be selected for immune therapies like anti-PD1. This prognostic model can be used ti identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with colon cancer that also involves the use of a composite model to predict the risk of death. This method also involves first determining gene expression intensities for several signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is an immune signature gene score. This immune signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 12, or genes highly correlated to the mean log expression of genes in Table 12, such as IKZF1, ITGAL, CD2, ITK, MAP4K1, CD3E, TBC1D10C, TRBC2, CD247, and CD3D. In some embodiments, one of the components is a hypoxia signature gene score. This hypoxia signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 13, or genes highly correlated to the mean log expression of genes in Table 13, such as SLC2A1, RALA, ERO1L, ANLN, S100A2, PHLDA2, CDC20, LAMC2, PLAUR, and SLC16A3. In some embodiments, one of the components is a vimentin (VIM) correlated gene score. This VIM correlated gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 14, or genes highly correlated to the mean log expression of genes in Table 14, such as CCDC80, VIM, HEG1, CNRIP1, RAB31, EFEMP2, GNB4, MRAS, CMTM3, and TIMP2. In some embodiments, one of the components is a CDH1 correlated gene score. This CDH1 correlated gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 15, or genes highly correlated to the mean log expression of genes in Table 15, such as ELF3, CLDN7, CLDN4, CDH1, RAB25, ESRP1, ESRP2, ERBB3, AP1M2, and EPCAM. In some embodiments, one of the components is a first prognosis signature gene score. This first prognosis signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 16, or genes highly correlated to the mean log expression of genes in Table 16, such as MZB1, OR6C4IGKV3-11 IGKV3D-11 IGKV3D-20 RHNO1, TNFRSF17, IGKC IGKV1D-39 IGKV1-39, IGHG1 IGH, IGLC1, IGKC IGKV1-16 IGKV1D-16, IGLV6-57, IGLV1-40 IGLV5-39, and IGJ. In some embodiments, one of the components is a second prognosis signature gene score. This second prognosis signature gene score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 17, or genes highly correlated to the mean log expression of genes in Table 17, such as SPP1, CDH2, ITGB1, SERPINE1, PLOD2, COL4A1, NTM, MPRIP, PLIN2, and TIMP1. The method can further involve determining the composite tumor stage. The method can then involve calculating a colon cancer risk score from the gene expression intensities of each category and the composite tumor stage, e.g., such that a colon breast cancer risk score is an indication that the subject has a high risk of death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. For example, patients with high risk scores can be more aggressively treated with chemotherapies like 5_FU with leucovorin, or Camptosar and Eloxatin, or combinations. These patients could also be preferentially considered for genetic tests for targeted therapies like EGFR and VEGF inhibitors. Patients with high immune signatures could be selected for immune therapies like anti-PD1. This prognostic model can be used to identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with kidney cancer that involves the use of correlated and anti-correlated biomarkers to predict the risk of death. This method involves first determining gene expression intensities for two signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 22, or genes highly correlated to the mean log expression of genes in Table 22, such as CRY2, NR3C2, HLF, EMX2OS, FAM221B, BDH2, BCL2, ACADL, NDRG2, and NPR3. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 23, or genes highly correlated to the mean log expression of genes in Table 23, such as TPX2, CCNB2, AURKB, HJURP, CENPA, CENPF, SKA1, CEP55, PTTG1, and FOXM1. The method can then involve calculating a kidney cancer risk score from the gene expression intensities of each category, e.g., such that a high kidney cancer risk score is an indication that the subject has a high risk of death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. For example, patients with high risk scores can be more aggressively treated with immunotherapies and targeted with drugs like Sorafenib, Sunitinib, Tersirolimus, Everolimus, Avastin, Votrient, and Axitinib. This prognostic model can be used to identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with brain cancer that also involves the use of a composite model to predict the risk of death. This method also involves first determining gene expression intensities for several signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 26, or genes highly correlated to the mean log expression of genes in Table 26, such as HLF, CTBP2, CPEB3, SGMS1, CTBP2, ZRANB1, BTRC, ACADSB, ZC3H12B, and REPS2. In some embodiments, one of the components is a second prognosis signature score.

This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 27, or genes highly correlated to the mean log expression of genes in Table 27, such as SKA1, TPX2, CCNB2, CENPA, BIRC5, RRM2, AURKA, AURKB, KIF2C, and CDCA8. In some embodiments, one of the components is a hypoxia signature score. This hypoxia signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 28, or genes highly correlated to the mean log expression of genes in Table 28, such as TREM1, SERPINE1, HILPDA, RALA, AK2, SOD2, ARL4C, PGK1, ANGPTL4, and SLC16A3. The method can then involve calculating a brain cancer risk score from the gene expression intensities of each category, e.g., such that a high brain cancer risk score is an indication that the subject has a high risk of death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. For example, patients with high risk scores can be more aggressively treated with chemotherapies like cisplatin, carboplatin, methotrexate, or combinations. These patients could also be preferentially considered for genetic tests for targeted therapies like Avastin and Everolimus. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with prostate cancer that involves the use of correlated and anti-correlated biomarkers to predict the risk of death. This method involves first determining gene expression intensities for two signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 31, or genes highly correlated to the mean log expression of genes in Table 31, such as LMOD1, PGM5, MYLK, SYNPO2, SORBS1, PPP1R12B, DES, CNN1, MYH11, and MYOCD. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 32, or genes highly correlated to the mean log expression of genes in Table 32, such as TPX2, UBE2C, PTTG1, NUSAP1, CENPA, AURKA, CDCA5, NUSAP1, AURKB, and BIRC5. The method can then involve calculating a prostate cancer risk score from the gene expression intensities of each category, e.g., such that a high prostate cancer risk score is an indication that the subject has a high risk of death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. In general, prostate cancer patients have relatively good outcomes, so “watchful waiting” and hormonal therapies are common treatments for prostate cancer patients. However, patients with high risk scores have extremely poor outcome and should be treated aggressively by chemotherapies like docetaxel. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with pancreatic cancer that involves the use of correlated and anti-correlated biomarkers to predict the risk of death. This method involves first determining gene expression intensities for two signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 33, or genes highly correlated to the mean log expression of genes in Table 33, such as RUNDC3A, PCLO, SVOP, CELF4, CPLX2, SCG3, DNAJC6, AP3B2, SCN3B, and MPP2. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 33, or genes highly correlated to the mean log expression of genes in Table 33, such as SFN, LAMB3, TMPRSS4, PLEK2, MST1R, GJB3, S100A16, GPRC5A, PLAUR, and CAPG. The method can then involve calculating a pancreatic cancer risk score from the gene expression intensities of each category, e.g., such that a high pancreatic cancer risk score is an indication that the subject has a high risk of death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. In general, pancreatic cancer patients have very poor outcomes and should be treated aggressively. However, patients with low risk scores have good outcome and could be considered for less toxic treatments. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with endometrium cancer that involves the use of correlated and anti-correlated biomarkers to predict the risk of death. This method involves first determining gene expression intensities for two signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 35, or genes highly correlated to the mean log expression of genes in Table 35, such as PGR, UBXN10, SNTN, SPATA18, VWA3A, CDHR4, WDR96, STX18, ARMC3, and ESR1. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 36, or genes highly correlated to the mean log expression of genes in Table 36, such as MRGBP, UBE2S, GMPS, ACOT7, E2F1, CENPO, MRGBP, AURKA, BIRC5, and TPX2. The method can then involve calculating a endometrium cancer risk score from the gene expression intensities of each category, e.g., such that a high endometrium cancer risk score is an indication that the subject has a high risk of death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. In general, endometrium cancer patients have very poor outcomes and should be treated aggressively with chemo- and radiation-therapy. However, patients with low risk scores have good outcome and could be considered for less toxic treatments, like hormonal therapy. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with melanoma that involves the use of correlated and anti-correlated biomarkers to predict the risk of death. This method involves first determining gene expression intensities for two signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 37, or genes highly correlated to the mean log expression of genes in Table 37, such as IKZF3, CD3G, SH2D1A, SLAMF6, CD247, SLAMF6, SIRPG, TRAF3IP3, THEMIS, and TBC1D10C. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 38, or genes highly correlated to the mean log expression of genes in Table 38, such as ITFG3, TMEM201, TBC1D16, PPT2, GCAT, PAK4, OTUD7B, FITM2, PCGF2, and GCAT. The method can then involve calculating a melanoma risk score from the gene expression intensities of each category, e.g., such that a high melanoma risk score is an indication that the subject has a high risk of death. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. In general, melanoma patients have very poor outcomes and should be treated aggressively. However, patients with low risk scores have good outcome and could be considered for less toxic treatments. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy. One of the prognostic signatures is immune signature, and high immune signature score is correlated with good outcome, so the low risk score can also be used to select patients for immunotherapies like PD-1, PDL1 and CTLA4 antibodies. The melanoma prognosis model can also predict outcome of non-melanoma skin cancer patients.

Also disclosed is a method for predicting prognosis of a patient with soft tissue cancer that involves the use of correlated and anti-correlated biomarkers to predict the risk of death. This method involves first determining gene expression intensities for signature genes components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a proliferation signature score. This proliferation signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 44, or genes highly correlated to the mean log expression of genes in Table 44, such as TPX2, CCNB2, CENPA, SKA1, CCNB1, KIF2C, CDCA8, DEPDC1, CDCA5, BIRC5. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 40, or genes highly correlated to the mean log expression of genes in Table 40, such as EFCAB14, RGS5, EPS15, EFCAB14, IL33, SNRK, FBXL3, MBNL1, HIPK3, and CMAHP. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 41, or genes highly correlated to the mean log expression of genes in Table 41, such as MRPS12, ALYREF, SNRPB, LSM12, UBE2S, BANF1, LSM4, ANAPC11, HNRNPK, and RANBP1. The method can then involve calculating a soft tissue cancer risk score from the gene expression intensities of one or more of these components, e.g., such that a high soft tissue cancer risk score is an indication that the subject has a high risk of death. Treatment of soft tissue cancers includes surgery, radiation, chemo- and targeted therapies. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. In general, soft tissue cancer patients have very poor outcomes and should be treated aggressively, including combinations of therapies. However, patients with low risk scores have good outcome and could be considered for less toxic treatments. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with uterine cancer that involves the use of correlated and anti-correlated biomarkers to predict the risk of death. This method involves first determining gene expression intensities for two signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 47, or genes highly correlated to the mean log expression of genes in Table 47, such as KIAA1324, CAPS, SCGB2A1, UBXN10, SOX17, RNF183, ASRGL1, UBXN10, SCGB1D2, and SPDEF. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 48, or genes highly correlated to the mean log expression of genes in Table 48, such as MRGBP, NUP155, GMPS, RYR1, FANCE, RFC4, UBE2S, ZNF623, ACOT7, and UCHL1. The method can then involve calculating a uterine cancer risk score from the gene expression intensities of each category, e.g., such that a high uterine cancer risk score is an indication that the subject has a high risk of death. The treatments to uterine cancer include surgery, radiation, hormonal (progestin) and chemotherapy. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. In general, uterine cancer patients have very poor outcomes and should be treated aggressively, including combinations of therapies like hormonal+chemotherapies. However, patients with low risk scores have good outcome and could be considered for less toxic treatments like hormonal (progestin) only. Hormonal receptors like PGR and ESR1 are highly expressed in relative lower risk patients, making them a good target group for progestin treatment. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with ovarian cancer that involves stratification of patients using signature score by genes in Table 51, and then the use of correlated and anti-correlated biomarkers to predict the risk of death in the “signature-low” group. This method involves first determining gene expression intensities for two signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 52, or genes highly correlated to the mean log expression of genes in Table 52, such as WDR96, DNAH6, TSNAXIP1, DNAH7, TTC18, PIFO, TTC25, NME5, WDR78, and DNAAF1. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 53, or genes highly correlated to the mean log expression of genes in Table 53, such as SPHK1, LINC00607, TNFAIP6, FAP, PTGIR, PLAU, TIMP3, INHBA, GPR68, and NTM. The method can then involve calculating an ovarian cancer risk score from the gene expression intensities of each category, e.g., such that a high ovarian cancer risk score is an indication that the subject has a high risk of death. The treatments for ovarian cancer include surgery and chemotherapy (platinum based and non-platinum based). The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. In general, ovarian cancer patients have very poor outcomes and should be treated aggressively. However, patients with low risk scores have good outcome and could be considered for less toxic treatments. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

Also disclosed is a method for predicting prognosis of a patient with bladder cancer that involves the use of correlated and anti-correlated biomarkers to predict the risk of death. This method involves first determining gene expression intensities for two signature gene components from a tumor biopsy sample from the subject. In some embodiments, one of the components is a first prognosis signature score. This first prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 57, or genes highly correlated to the mean log expression of genes in Table 57, such as ITGAL, IKZF1, CD3E, CD48, SLAMF6, CD2, TBC1D10C, PVRIG, CD5, and SLA2. In some embodiments, one of the components is a second prognosis signature score. This second prognosis signature score can be generated using at least 1, 2, 3, 4, 5 6, 7, 8, 9, or 10 of the genes listed in Table 58, or genes highly correlated to the mean log expression of genes in Table 58, such as KRT6B, DSC2, DSG3, FAM106B, KRT6A, KRT14, SPRR2D, RALA, SERPINB5, and RHCG. The method can then involve calculating bladder cancer risk score from the gene expression intensities of each category, e.g., such that a high bladder cancer risk score is an indication that the subject has a high risk of death. Treatment options for bladder cancer include surgery, radiation, chemo- and immune-therapies. The method can further involve treating the subject with more aggressive treatment if the subject has a high risk score. In general, bladder cancer patients have very poor outcomes and should be treated aggressively. However, patients with low risk scores have good outcome and could be considered for less toxic treatments, like immune therapies. One signature component is immune signature, and high immune signature is correlated with relatively good outcome. This suggests low-risk bladder patients are immune therapy target group. This prognostic model can be used for identify patients with unmet medical needs for new clinical trials for pharmaceutical companies, and to match case and control groups with similar prognostic levels for better clinical trial design for treatment efficacy.

In each of the above methods, risk scores can be calculate by any suitable computational predictive model, such as general linear regression, logistic regression, or simple linear/non-linear multivariate models with equal or unequal contributions from each component. In some case, the method involves simply summing the number of risk factors.

The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.

DESCRIPTION OF DRAWINGS

FIG. 1 is a graph showing that a 5-component model predicts average patient death rate in the validation set of primary breast cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 100 patients as ranked by the prediction.

FIG. 2 is a graph showing that the survival model predicts average bone metastasis rate in validation set of patients with primary tumor. X-axis: predicted death rate. Y-axis: average bone metastasis rate (running average of 100 samples ranked by predicted score).

FIG. 3 shows Kaplan-Meier plots for 1249 primary breast cancer patients in the validation set. Top curve: prediction score<0.15, Middle curve: score between 0.2 and 0.35, Bottom curve: score>0.35. The P-value for the Chi-square test is 0.

FIG. 4 is a graph showing that a 6-component model predicts average patient death rate in the validation set of lung cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 200 patients as ranked by the prediction.

FIG. 5 shows Kaplan-Meier plots for 1168 lung cancer patients in the validation set. Top curve: risk score<0.4, Middle curve: score between 0.4 and 0.7, Bottom curve: score>0.7. The P-value for the Chi-square test is 0.

FIG. 6 is a graph showing a 5-component model (based on reduced gene sets) predicts average patient death rate in the validation set of lung cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 200 patients as ranked by the prediction.

FIG. 7 shows Kaplan-Meier plots for 1168 lung cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.4, Middle curve: score between 0.4 and 0.7, Bottom curve: score>0.7. The P-value for the Chi-square test is 0.

FIG. 8 is a graph showing microarray components (without tumor stage) predict average patient death rate in the validation set of lung cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 200 patients as ranked by the prediction.

FIG. 9 is a graph showing an 8-component model predicts average patient death rate in the validation set of colon cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 200 patients as ranked by the prediction.

FIG. 10 shows Kaplan-Meier plots for 1057 colon cancer patients in the validation set. Top curve: risk score<0.2, Middle curve: score between 0.2 and 0.5, Bottom curve: score>0.5. The P-value for the Chi-square test is 3.86×10−12.

FIG. 11 is a graph showing a 7-component model predicts average patient death rate in colon cancer patients (based on reduced gene sets). X-axis: predicted death rate, Y-axis: actual average death rate, running average of 200 patients as ranked by the prediction.

FIG. 12 shows Kaplan-Meier plots for 1057 colon cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.25, Middle curve: score between 0.25 and 0.5, Bottom curve: score>0.5. The P-value for the Chi-square test is 3.7×10−13.

FIG. 13 is a graph showing microarray components (without tumor stage) predict average patient death rate in colon cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 200 patients as ranked by the prediction.

FIG. 14 is a graph showing a 2-component model predicts average patient death rate in validation set of kidney cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 100 patients as ranked by the prediction.

FIG. 15 shows Kaplan-Meier plots for 444 kidney cancer patients in the validation set. Top curve: risk score<0.35, Middle curve: score between 0.35 and 0.6, Bottom curve: score>0.6. The P-value for the Chi-square test is 2.4×10−14. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 16 is a graph showing a 2-component model predicts average patient death rate in kidney cancer patients (based on reduced gene sets). X-axis: predicted death rate, Y-axis: actual average death rate, running average of 100 patients as ranked by the prediction.

FIG. 17 shows Kaplan-Meier plots for 444 kidney cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.35, Middle curve: score between 0.35 and 0.6, Bottom curve: score>0.6. The P-value for the Chi-square test is 1.4×10−15. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 18 is a graph showing a 3-component model predicts average patient death rate in the validation set of brain cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 100 patients as ranked by the prediction.

FIG. 19 shows Kaplan-Meier plots for 257 brain cancer patients in the validation set. Top curve: risk score<0.4, Middle curve: score between 0.4 and 0.75, Bottom curve: score>0.75. The P-value for the Chi-square test is 3.2×10−13. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group)

FIG. 20 is a graph showing a 3-component model predicts average patient death rate in brain cancer patients (based on reduced gene sets). X-axis: predicted death rate, Y-axis: actual average death rate, running average of 100 patients as ranked by the prediction.

FIG. 21 shows Kaplan-Meier plots for 257 brain cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.4, Middle curve: score between 0.4 and 0.75,

Bottom curve: score>0.75. The P-value for the Chi-square test is 6.8×10−13. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 22 is a Kaplan-Meier plots for 151 prostate cancer patients in the validation set. Top curve: risk score<0.4, Bottom curve: score>0.4. The P-value for the Chi-square test is 0. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 23 is a Kaplan-Meier plots for 151 prostate cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.4, Bottom curve: score>0.4. The P-value for the Chi-square test is 0. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 24 shows Kaplan-Meier plots for 263 pancreatic cancer patients in the validation set. Top curve: risk score<0.5, Bottom curve: score>0.5. The P-value for the Chi-square test is 5.82×10−9. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 25 shows Kaplan-Meier plots for 263 pancreatic cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.5, Bottom curve: score>0.5. The P-value for the Chi-square test is 3.8×10−8. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group.

FIG. 26 is a plot showing a 3-component model predicts average patient death rate in the validation set of endometrium cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 27 shows Kaplan-Meier plots for 184 endometrium cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.2, Middle curve: score between 0.2 and 0.4, Bottom curve: score>0.4. The P-value for the Chi-square test is 9.7×10−5.

FIG. 28 shows Kaplan-Meier plots for 184 endometrium cancer patients in the validation set. Top curve: risk score<0.2, Middle curve: score between 0.2 and 0.4, Bottom curve: score >0.4. The P-value for the Chi-square test is 1.0×10−4.

FIG. 29 is a plot showing a 2-component model predicts average patient death rate in the validation set melanoma patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 30 shows Kaplan-Meier plots for 153 melanoma patients in the validation set. Top curve: risk score<0.45, Middle curve: score between 0.45 and 0.65, Bottom curve: score>0.65. The P-value for the Chi-square test is 9.3×10−9.

FIG. 31 is a plot showing a 2-component model predicts average patient death rate in melanoma patients (based on reduced gene sets). X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 32 shows Kaplan-Meier plots for 153 melanoma patients in the validation set (based on reduced gene sets). Top curve: risk score<0.45, Middle curve: score between 0.45 and 0.6, Bottom curve: score>0.6. The P-value for the Chi-square test is 1.0×10−7.

FIG. 33 shows Kaplan-Meier plots for 152 other skin cancer patients excluding malignant melanoma. Top curve: risk score<0.45, Middle curve: score between 0.45 and 0.6, Bottom curve: score>0.6. The P-value for the Chi-square test is 9.2×10−4.

FIG. 34 is a graph showing a 2-component model predicts average patient death rate in the validation set of soft tissue cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 35 shows Kaplan-Meier plots for 95 soft tissue cancer patients in the validation set. Top curve: risk score<0.34, Middle curve: score between 0.34 and 0.55, Bottom curve: score >0.55. The P-value for the Chi-square test is 1.1×10−4. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 36 shows Kaplan-Meier plots for 95 soft tissue cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.34, Middle curve: score between 0.34 and 0.55, Bottom curve: score>0.55. The P-value for the Chi-square test is 3.2×10−4. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 37 is a plot showing model based on proliferation signature predicts average patient death rate in soft tissue cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 38 shows Kaplan-Meier plots based on proliferation signature for 95 soft tissue cancer patients in the validation set. Top curve: risk score<0.42, Middle curve: score between 0.42 and 0.55, Bottom curve: score>0.55. The P-value for the Chi-square test is 2.3×10−4. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 39 shows Kaplan-Meier plots for 95 soft tissue cancer patients in the validation set (based on reduced proliferation geneset). Top curve: risk score<0.4, Middle curve: score between 0.4 and 0.55, Bottom curve: score>0.55. The P-value for the Chi-square test is 1.2×10−4. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 40 shows Kaplan-Meier plots for 95 soft tissue cancer patients in the validation set, by the average risk score. Top curve: risk score<0.4, Middle curve: score between 0.4 and 0.55, Bottom curve: score>0.55. The P-value for the Chi-square test is 1.2×10−4. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 41 shows Kaplan-Meier plots for 95 soft tissue cancer patients in the validation set, by the number of risk factors (RF). Top curve: RF =0, Middle RF =1, Bottom curve: RF =2. The P-value for the Chi-square test is 5.7×10−5. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group).

FIG. 42 is a plot showing a 3-component model predicts average patient death rate in the validation set of uterus cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 43 shows Kaplan-Meier plots for 153 uterus cancer patients in the validation set. Top curve: risk score<0.32, Middle curve: score between 0.32 and 0.6, Bottom curve: score>0.6. The P-value for the Chi-square test is 2.1×10−9.

FIG. 44 is a plot showing a 3-component model predicts average patient death rate in uterus cancer patients (based on reduced gene sets). X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 45 shows Kaplan-Meier plots for 153 uterus cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.32, Middle curve: score between 0.32 and 0.6, Bottom curve: score>0.6. The P-value for the Chi-square test is 1.3×10−9.

FIG. 46 is a histogram of X2 intensities (average of log2 intensities from all probes in Table 51).

FIG. 47 is a plot showing estrogen-receptor (ER) intensity vs. X2 intensity. High-X2 patients have uniform high ER levels.

FIG. 48 is a plot showing a 3-component model predicts average patient death rate in X2-ovarian cancer patients. X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 49 shows Kaplan-Meier plots for 170 X2-ovarian cancer patients in the validation set. Top curve: risk score<0.5, Middle curve: score between 0.5 and 0.7, Bottom curve: score >0.7. The P-value for the Chi-square test is 3.6×10−7. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group.

FIGS. 50A and 50B show Kaplan-Meier plots for signatures (FIG. 50A) and tumor stage (FIG. 50B) in 170 X2-ovarian cancer patients of the validation set. In FIG. 50A, Top curve: risk score<0, Middle curve: score between 0 and 0.2, Bottom curve: score>0.2. The Chi-square for 2 degree of freedom is 34. In FIG. 50B, Top curve: tumor stage 0, 1, 2; Middle curve: tumor stage 3; Bottom curve: tumor stage 4. The Chi-square for 2 degree of freedom is 27.9.

FIG. 51 is a plot showing a 3-component model predicts average patient death rate in X2-ovarian cancer patients (based on reduced gene sets). X-axis: predicted death rate, Y-axis: actual average death rate, running average of 50 patients as ranked by the prediction.

FIG. 52 shows Kaplan-Meier plots for 170 X2-ovarian cancer patients in the validation set. Top curve: risk score<0.5, Middle curve: score between 0.5 and 0.7, Bottom curve: score >0.7. The P-value for the Chi-square test is 2.1×10−7. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group.

FIGS. 53A and 53B are histograms of immune signature score for X2-(FIG. 53A) and X2+ (FIG. 53B) patients.

FIG. 54 shows the correlation between CDH6 and X2 (correlation=0.61).

FIGS. 55A and 55B are Kaplan-Meier curves for X2-population (FIG. 55A) and X2+ population (FIG. 55B).

FIG. 56 shows Kaplan-Meier plots for 136 bladder cancer patients in the validation set. Top curve: risk score<0.66, Middle curve: score between 0.66 and 0.75, Bottom curve: score >0.75. The P-value for the Chi-square test is 1.3×10−3. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group.

FIG. 57 shows Kaplan-Meier plots for 136 bladder cancer patients in the validation set (based on reduced gene sets). Top curve: risk score<0.5, Middle curve: score between 0.5 and 0.75, Bottom curve: score>0.75. The P-value for the Chi-square test is 2.2×10−3. Note the K-M curves are biased given significant number of follow-up dates are missing for the good outcome patients. The chi-square test p-value is still correct since it only uses live/death information in each group.

DETAILED DESCRIPTION

Prognostic and predictive biomarkers are disclosed that can be used in systems and methods for predicting the prognosis of a cancer patient, which can be used to guide therapeutic and palliative treatment of the patient. The methods generally involve determining gene expression of a panel of biomarkers and use of these gene expression intensities calculate predictive risk scores.

Gene Expression Assays

Methods of “determining gene expression levels” include methods that quantify levels of gene transcripts as well as methods that determine whether a gene of interest is expressed at all. A measured expression level may be expressed as any quantitative value, for example, a fold-change in expression, up or down, relative to a control gene or relative to the same gene in another sample, or a log ratio of expression, or any visual representation thereof, such as, for example, a “heat crap” where a color intensity is representative of the amount of gene expression detected. Exemplary methods for detecting the level of expression of a gene include, but are not limited to, Northern blotting, dot or slot blots, reporter gene matrix, nuclease protection, RT-PCR, microarray profiling, differential display, 2D gel electrophoresis, SELDI-TOF, ICAT, enzyme assay, antibody assay, and MNAzyme-based detection methods. Optionally a gene whose level of expression is to be detected may be amplified, for example by methods that may include one or more of: polymerase chain reaction (PCR), strand displacement amplification (SDA), loop-mediated isothermal amplification (LAMP), rolling circle amplification (RCA), transcription-mediated amplification (TMA), self-sustained sequence replication (3SR), nucleic acid sequence based amplification (NASBA), or reverse transcription polymerase chain reaction (RT- PCR).

A number of suitable high throughput formats exist for evaluating expression patterns and profiles of the disclosed genes. Numerous technological platforms for performing high throughput expression analysis are known. Generally, such methods involve a logical or physical array of either the subject samples, the biomarkers, or both. Common array formats include both liquid and solid phase arrays. For example, assays employing liquid phase arrays, e.g., for hybridization of nucleic acids, binding of antibodies or other receptors to ligand, etc., can be performed in multiwell or microtiter plates. Microtiter plates with 96, 384 or 1536 wells are widely available, and even higher numbers of wells, e.g., 3456 and 9600 can be used. In general, the choice of microtiter plates is determined by the methods and equipment, e.g., robotic handling and loading systems, used for sample preparation and analysis. Exemplary systems include, e.g., xMAP® technology from Luminex (Austin, Tex.), the SECTOR® Imager with MULTI-ARRAY® and MULTI-SPOT® technologies from Meso Scale Discovery (Gaithersburg, Md.), the ORCA™ system from Beckman-Coulter, Inc. (Fullerton, Calif.) and the ZYMATETM systems from Zymark Corporation (Hopkinton, Mass.), miRCURY LNA™ microRNA Arrays (Exiqon, Woburn, Mass.).

Alternatively, a variety of solid phase arrays can favorably be employed to determine expression patterns in the context of the disclosed methods, assays and kits. Exemplary formats include membrane or filter arrays (e.g., nitrocellulose, nylon), pin arrays, and bead arrays (e.g., in a liquid “slurry”). Typically, probes corresponding to nucleic acid or protein reagents that specifically interact with (e.g., hybridize to or bind to) an expression product corresponding to a member of the candidate library, are immobilized, for example by direct or indirect cross-linking, to the solid support. Essentially any solid support capable of withstanding the reagents and conditions necessary for performing the particular expression assay can be utilized. For example, functionalized glass, silicon, silicon dioxide, modified silicon, any of a variety of polymers, such as (poly)tetrafluoroethylene, (poly)vinylidenedifluoride, polystyrene, polycarbonate, or combinations thereof can all serve as the substrate for a solid phase array.

In one embodiment, the array is a “chip” composed, e.g., of one of the above-specified materials. Polynucleotide probes, e.g., RNA or DNA, such as cDNA, synthetic oligonucleotides, and the like, or binding proteins such as antibodies or antigen-binding fragments or derivatives thereof, that specifically interact with expression products of individual components of the candidate library are affixed to the chip in a logically ordered manner, i.e., in an array. In addition, any molecule with a specific affinity for either the sense or anti-sense sequence of the marker nucleotide sequence (depending on the design of the sample labeling), can be fixed to the array surface without loss of specific affinity for the marker and can be obtained and produced for array production, for example, proteins that specifically recognize the specific nucleic acid sequence of the marker, ribozymes, peptide nucleic acids (PNA), or other chemicals or molecules with specific affinity.

Microarray expression may be detected by scanning the microarray with a variety of laser or CCD-based scanners, and extracting features with numerous software packages, for example, IMAGENE™ (Biodiscovery), Feature Extraction Software (Agilent), SCANLYZE™ (Stanford Univ., Stanford, Calif.), GENEPIX™ (Axon Instruments).

In some cases, single molecule sequencing methods are used determining gene expression patterns. In some embodiments, amplified cDNA is sequenced by whole transcriptome shotgun sequencing (also referred to herein as (“RNA-Seq”). Whole transcriptome shotgun sequencing (RNA-Seq) can be accomplished using a variety of next-generation sequencing platforms such as the Illumina Genome Analyzer platform, ABI Solid Sequencing platform, or Life Science's 454 Sequencing platform.

In some embodiments, the nCounter® Analysis system (Nanostring Technologies, Seattle, Wash.) is used to detect intrinsic gene expression. This system is described in International Patent Application Publication No. WO 08/124,847 and U.S. Pat. No. 8,415,102, which are each incorporated herein by reference in their entireties for the teaching of this system. The basis of the nCounter® Analysis system is the unique code assigned to each nucleic acid target to be assayed.

The code is composed of an ordered series of colored fluorescent spots which create a unique barcode for each target to be assayed. A pair of probes is designed for each DNA or RNA target, a biotinylated capture probe and a reporter probe carrying the fluorescent barcode. This system is also referred to, herein, as the nanoreporter code system.

Specific reporter and capture probes can be synthesized for each target. Briefly, sequence-specific DNA oligonucleotide probes are attached to code-specific reporter molecules. Preferably, each sequence specific reporter probe comprises a target specific sequence capable of hybridizing to no more than one target and optionally comprises at least two, at least three, or at least four label attachment regions, said attachment regions comprising one or more label monomers that emit light. Capture probes are made by ligating a second sequence-specific DNA oligonucleotide for each target to a universal oligonucleotide containing biotin. Reporter and capture probes are all pooled into a single hybridization mixture, the “probe library”.

The relative abundance of each target is measured in a single multiplexed hybridization reaction. The method comprises contacting a biological sample with a probe library, the library comprising a probe pair for gene target, such that the presence of the target in the sample creates a probe pair target complex. The complex is then purified. More specifically, the sample is combined with the probe library, and hybridization occurs in solution. After hybridization, the tripartite hybridized complexes (probe pairs and target) are purified in a two-step procedure using magnetic beads linked to oligonucleotides complementary to universal sequences present on the capture and reporter probes, This dual purification process allows the hybridization reaction to be driven to completion with a large excess of target-specific probes, as they are ultimately removed, and, thus, do not interfere with binding and imaging of the sample. All post hybridization steps are handled robotically on a custom liquid-handling robot (Prep Station, NanoString Technologies).

Purified reactions are deposited by the Prep Station into individual flow cells of a sample cartridge, bound to a streptavidin-coated surface via the capture probe, electrophoresed to elongate the reporter probes, and immobilized. After processing, the sample cartridge is transferred to a fully automated imaging and data collection device (Digital Analyzer, NanoString Technologies). The expression level of a target is measured by imaging each sample and counting the number of times the code for that target is detected. Data is output in simple spreadsheet format listing the number of counts per target, per sample.

This system can be used along with nanoreporters. Additional disclosure regarding nanoreporters can be found in International Publication No. WO 07/076,129 and WO 07/076,132, and US Patent Publication No. 2010/0015607 and 2010/0261026, the contents of which are incorporated herein in their entireties. Further, the term nucleic acid probes and nanoreporters can include the rationally designed (e.g, synthetic sequences) described in International Publication No. WO 2010/019826 and US Patent Publication No. 2010/0047924, incorporated herein by reference in its entirety.

Calculation of Risk Score

From the disclosed gene expression values, a dataset can be generated and inputted into an analytical classification process that uses the data to classify the biological sample with a risk score. The data may be obtained via any technique that results in an individual receiving data associated with a sample. For example, an individual may obtain the dataset by generating the dataset himself by methods known to those in the art. Alternatively, the dataset may be obtained by receiving a dataset or one or more data values from another individual or entity. For example, a laboratory professional may generate certain data values while another individual, such as a medical professional, may input all or part of the dataset into an analytic process to generate the result.

Prior to input into the analytical process, the data in each dataset can be collected by measuring the values for each biomarker gene, usually in duplicate or triplicate or in multiple replicates. The data may be manipulated, for example raw data may be transformed using standard curves, and the average of replicate measurements used to calculate the average and standard deviation for each patient. These values may be transformed before being used in the models.

For example, it is often useful to pre-process gene expression data, for example, by addressing missing data, translation, scaling, normalization, weighting, etc. Multivariate projection methods, such as principal component analysis (PCA) and partial least squares analysis (PLS), are so-called scaling sensitive methods. By using prior knowledge and experience about the type of data studied, the quality of the data prior to multivariate modeling can be enhanced by scaling and/or weighting. Adequate scaling and/or weighting can reveal important and interesting variation hidden within the data, and therefore make subsequent multivariate modeling more efficient. Scaling and weighting may be used to place the data in the correct metric, based on knowledge and experience of the studied system, and therefore reveal patterns already inherently present in the data.

If possible, missing data, for example gaps in column values, should be avoided. However, if necessary, such missing data may replaced or “filled” with, for example, the mean value of a column (“mean fill”); a random value (“random fill”); or a value based on a principal component analysis (“principal component fill”). In some cases, there are multiple genes from the same pathway signature, and the missing data of a particular genes can be modeled by correlated genes in the same pathway.

“Translation” of the descriptor coordinate axes can be useful. Examples of such translation include normalization and mean centering. “Normalization” may be used to remove sample-to-sample variation. Some commonly used methods for calculating normalization factor include: (i) global normalization that uses all genes on the array; (ii) housekeeping genes normalization that uses constantly expressed housekeeping/invariant genes; and (iii) internal controls normalization that uses known amount of exogenous control genes added during hybridization. In some embodiments, the intrinsic genes disclosed herein can be normalized to control housekeeping genes. It will be understood by one of skill in the art that the methods disclosed herein are not bound by normalization to any particular housekeeping genes, and that any suitable housekeeping gene(s) known in the art can be used.

Many normalization approaches are possible, and they can often be applied at any of several points in the analysis. In one embodiment, data is normalized using the LOWESS method, which is a global locally weighted scatter plot smoothing normalization function. In another embodiment, data is normalized to the geometric mean of set of multiple housekeeping genes.

“Mean centering” may also be used to simplify interpretation. Usually, for each descriptor, the average value of that descriptor for all samples is subtracted. In this way, the mean of a descriptor coincides with the origin, and all descriptors are “centered” at zero. In “unit variance scaling,” data can be scaled to equal variance. Usually, the value of each descriptor is scaled by 1/StDev, where StDev is the standard deviation for that descriptor for all samples. “Pareto scaling” is, in some sense, intermediate between mean centering and unit variance scaling. In pareto scaling, the value of each descriptor is scaled by 1/sqrt(StDev), where StDev is the standard deviation for that descriptor for all samples. In this way, each descriptor has a variance numerically equal to its initial standard deviation. The pareto scaling may be performed, for example, on raw data or mean centered data.

“Logarithmic scaling” may be used to assist interpretation when data have a positive skew and/or when data spans a large range, e.g., several orders of magnitude. Usually, for each descriptor, the value is replaced by the logarithm of that value. In “equal range scaling,” each descriptor is divided by the range of that descriptor for all samples. In this way, all descriptors have the same range, that is, 1. However, this method is sensitive to presence of outlier points. In “autoscaling,” each data vector is mean centered and unit variance scaled. This technique is a very useful because each descriptor is then weighted equally, and large and small values are treated with equal emphasis. This can be important for genes expressed at very low, but still detectable, levels.

Data can also be normalized by the method described by Welsh et al. BMC Bioinformatics. 2013 14:153, which is incorporated by reference for its teaching of these algorithms and methods.

The methods described herein may be implemented and/or the results recorded using any device capable of implementing the methods and/or recording the results. Examples of devices that may be used include but are not limited to electronic computational devices, including computers of all types. When the methods described herein are implemented and/or recorded in a computer, the computer program that may be used to configure the computer to carry out the steps of the methods may be contained in any computer readable medium capable of containing the computer program. Examples of computer readable medium that may be used include but are not limited to diskettes, CD-ROMs, DVDs, ROM, RAM, and other memory and computer storage devices. The computer program that may be used to configure the computer to carry out the steps of the methods and/or record the results may also be provided over an electronic network, for example, over the internet, an intranet, or other network.

This data can then be input into the analytical process with defined parameter, The analytic classification process may be any type of learning algorithm with defined parameters, or in other words, a predictive model. In general, the analytical process will be in the form of a model generated by a statistical analytical method such as those described below. Examples of such analytical processes may include a linear algorithm, a quadratic algorithm, a polynomial algorithm, a decision tree algorithm, or a voting algorithm.

Using any suitable learning algorithm, an appropriate reference or training dataset can be used to determine the parameters of the analytical process to be used for classification, i.e., develop a predictive model. The reference or training dataset to be used will depend on the desired classification to be determined, The dataset may include data from two, three, four or more classes,

The number of features that may be used by an analytical process to classify a test subject with adequate certainty is 2 or more, in some embodiments, it is 3 or more, 4 or more, 10 or more, or between 10 and 74. Depending on the degree of certainty sought, however, the number of features used in an analytical process can be more or less, but in all cases is at least 2. In one embodiment, the number of features that may be used by an analytical process to classify a test subject is optimized to allow a classification of a test subject with high certainty.

Suitable data analysis algorithms are known in the art. In one embodiment, a data analysis algorithm of the disclosure comprises Classification and Regression Tree (CART), Multiple Additive Regression Tree (MART), Prediction Analysis for Microarrays (PAM), or Random Forest analysis. Such algorithms classify complex spectra from biological materials to distinguish subjects as normal or as possessing biomarker levels characteristic of a particular disease state. In other embodiments, a data analysis algorithm of the disclosure comprises ANOVA and nonparametric equivalents, linear discriminant analysis, logistic regression analysis, nearest neighbor classifier analysis, neural networks, principal component analysis, quadratic discriminant analysis, regression classifiers and support vector machines. While such algorithms may be used to construct an analytical process and/or increase the speed and efficiency of the application of the analytical process and to avoid investigator bias, one of ordinary skill in the art will realize that computer-based algorithms are not required to carry out the methods of the present disclosure.

As will be appreciated by those of skill in the art, a number of quantitative criteria can be used to communicate the performance of the comparisons made between a test marker profile and reference marker profiles. These include area under the curve (AUC), hazard ratio (HR), relative risk (RR), reclassification, positive predictive value (PPV), negative predictive value (NPV), accuracy, sensitivity and specificity, Net reclassification Index, Clinical Net reclassification Index. In addition, other constructs such a receiver operator curves (ROC) can be used to evaluate analytical process performance.

Predicting Cancer Survivability

The disclosed biomarkers, systems, methods, assays, and kits can be used to predict the survivability of a subject with a cancer. The disclosed biomarkers, methods, assays, and kits are particularly useful to predict the benefit of aggressive treatment. For example, the cancer of the disclosed methods can be any cell in a subject undergoing unregulated growth, invasion, or metastasis. In some aspects, the cancer can be any neoplasm or tumor for which radiotherapy is currently used. Alternatively, the cancer can be a neoplasm or tumor that is not sufficiently sensitive to radiotherapy using standard methods. Thus, the cancer can be a sarcoma, lymphoma, leukemia, carcinoma, blastoma, or germ cell tumor. A representative but non-limiting list of cancers that the disclosed compositions can be used to treat include lymphoma, B cell lymphoma, T cell lymphoma, mycosis fungoides, Hodgkin's Disease, myeloid leukemia, bladder cancer, brain cancer, nervous system cancer, head and neck cancer, squamous cell carcinoma of head and neck, kidney cancer, lung cancers such as small cell lung cancer and non-small cell lung cancer, neuroblastoma/glioblastoma, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, liver cancer, melanoma, squamous cell carcinomas of the mouth, throat, larynx, and lung, colon cancer, cervical cancer, cervical carcinoma, breast cancer, epithelial cancer, renal cancer, genitourinary cancer, pulmonary cancer, esophageal carcinoma, head and neck carcinoma, large bowel cancer, hematopoietic cancers; testicular cancer; colon and rectal cancers, prostatic cancer, and pancreatic cancer.

Adjuvant Therapy

The calculated risk scores can be used to predict the benefit of an adjuvant therapy for a subject based on their expected survivability. In some embodiments, the method also predicts the efficacy of adjuvant therapy in the subject. Adjuvant therapy is additional treatment given after surgery to reduce the risk that the cancer will come back. Adjuvant treatment may include chemotherapy (the use of drugs to kill cancer cells) and/or radiation therapy (the use of high energy x-rays to kill cancer cells).

The disclosed risk scores can be used to identify whether the subject will have improve survivability if treated with adjuvant chemotherapy (ACT) and may also predict benefit of radiation therapy. For example, the method can involve administering ACT and/or radiation therapy to the subject if a high risk score is calculated.

Definitions

The term “subject” refers to any individual who is the target of administration or treatment. The subject can be a vertebrate, for example, a mammal. Thus, the subject can be a human or veterinary patient. The term “patient” refers to a subject under the treatment of a clinician, e.g., physician.

The term “prognosis” refers to a predicted clinical outcome that can be used by a clinician to select an appropriate treatment. This term includes estimations of survival, tumor progression (e.g., metastasis), and/or responsiveness to treatment.

The term “treatment” refers to the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder. This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder.

A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.

EXAMPLES

Gene expression profiling data was generated for approximately 16,000 cancer subjects. This dataset is the biggest and one of the best quality dataset in the world. It was generated using a uniform protocol (NuGen) on a uniform platform (Merck version of Affymetrix® arrays).

The gene expression data in combination with patient clinical follow-up data (overall survival, response to standard care treatments, etc.) was used to discover prognostic or predictive biomarkers. There are more than 10 tumor types or subtypes with adequate number of samples to derive the prognosis signatures. For example, there are nearly 4,000 breast cancer samples, 500 brain tumors, 880 kidney tumors, 3,000 lung tumors and more than 2,000 colon tumors in the profiling dataset.

For those tumor types or subtypes with adequate number of samples, the approach for biomarker discovery was to divide the samples equally into two parts: the first half samples used for biomarker discovery and model training, and the second half used for validation.

Within the training samples, a modified method based on a previous publication (Dai H, et al. Cancer Res. 2005 65(10):4059-66) was used to discover two groups of biomarkers (correlated and anti-correlated to the survival). The mean log expression level of each biomarker group in each sample was computed, and the mean log expression of each group, or the difference of the mean log expression between these two groups of biomarkers was used to build a survival prediction model in the training samples. The same model was then applied to the reserved validation samples to estimate the performance.

For tumor-types with more than one or two mechanisms involved in affecting the final outcome, a composite model was developed to include these factors. For example, the factors can be pathway scores, single gene markers, or histo-pathological parameters.

Example 1 Prognostic Model for Breast Cancer

Proliferation is a strong predictor of metastasis or death in ER+breast cancer patients. Studies also linked estrogen receptor (ER) level and Her2 level to breast cancer patient outcome. In addition, it was observed in the dataset that the immune signature is related to good outcome in breast cancer patient, especially in ER-patients. For a strong predictor, all these factors can be included.

A composite model was therefore built in 2,000 breast cancer training samples. The model contained ER and HER2 expression levels as measured by array probes, average proliferation score measured by 100 proliferation genes, and immune score measured by 100 immune related genes.

The performance of this model was evaluated in reserved validation set of 2,000 samples. The validation set contains 1249 unique primary patients and 166 unique metastatic patients, with some samples profiled multiple times. FIG. 1 shows the predicted death rate vs. the actual average (running average of 100 samples as ranked by the prediction score) death rate in unique primaries. As shown in the Figure, the model predicts the average death rate very well.

The odds ratio in all 1,249 validation primary patients is 5.99, 95% CI [4.00, 8.98]. The predictor is independently predictive in each well define clinical sub-populations. In ER+patients, the odds ratio was 5.4, 95% CI [3.3, 8.9]. In ER-patients, the odds ratio was 4.8, 95% CI [2.2, 10.3]. In the metastatic population, the odds ratio was 8.4, 95% CI [3.1, 22.6].

This same model also predicts the bone metastasis in primary breast cancer patients. FIG. 2 shows the actual average bone metastasis rate vs. the predicted death rate. A strong correlation is observed between these two rates. Among 672 patients with low predicted score, 6 developed metastasis (0.9%), whereas in the 577 patients with high predicted score, 41 developed bone metastasis (7.1%), Fisher's exact test P-value is 4.2×10−9.

Based on the predictive score by the model, patients can be further divided into good (score <0.2), medium (0.2<score<0.35) and poor (score>0.35) prognosis groups. The actual death rates from the primary validation sets were 4.8% (32/672), 16.6% (62/373) and 34.8% (71/204).

In the validation set, there were 637 primary patients with lymph node negative (LNO) and 496 primary patients with lymph node positive (LN1, 2, 3) breast cancer. When the model was applied to the LN− and LN+positive groups, the odds ratios for the overall survival were 5.78, 95% CI[3.12, 10.69], and 5.06, 95% CI[2.54, 10.07] respectively. For the bone metastasis, in the LN−, the total bone metastasis rat is 1% (7/637), hence the prediction is not significant. In the LN+ group, the bone metastasis rates were 0.0% (0/179) and 9.8% (31/317), P-value=7.4×10−7.

When patients were divided up into age groups (less than 55 years and great than 55 years), the overall survival odds ratios were 9.15, 95% CI[3.57, 23.44], and 5.96, 95% CI[3.75, 9.45] respectively. The bone metastasis rates in the younger patient group were 1.9% (4/208) vs. 8.8% (23/261) for the low and high risk score groups (P=0.001). For the older patient group, the rates were 0.4% (2/464) vs. 5.7% (18/316), P-value=4.8×10−8.

When patients were divided into tumor grade groups 1&2, and 3, the overall survival odds ratios were 6.18 95% CI[3.78, 10.12] and 6.11, 95% CI[2.86, 13.07], respectively. In grade 1&2 patients, the bone metastasis rates were 0.4% (2/491) vs. 7.8% (22/282) for the low and high risk groups, P-value=1.6×10−8. For grade 3 patients, the rates were 2.2% (4/181) vs. 6.4% (19/295), P-value=0.05.

Materials & Methods

The 5 components used to determine a breast cancer risk score were: ER, measured by gene expression probe targeting NM_000125, in log2 scale; HER2, measured by gene expression probe, targeting NM_03_2339, in log2 scale; proliferation signature score, measured by mean log2 intensities of the genes in Table 1; immune signature score, measured by mean log2 intensities of the genes in Table 2; and composite stage based on histology and clinical stage.

The formulas used for calculating the breast prediction score were:


Breast Cancer Risk Score=0.653031+(−0.027485*ER)+(0.004901*HER2)+(0.047574*Proliferation)+(−0.071552*immune)   (Formula 1a),

where a high score means high risk.


Breast Cancer Risk Score=0.546072+(−0.025403*ER)+(−0.004187*HER2)+(0.042013*Proliferation)+(−0.073342*immune)+(0.126162*stage)   (Formula 1b),

where a high score means high risk.

TABLE 1 100 Proliferation genes Probe Gene merck-CR596700_a_at RRM2 merck2-AL517462_s_at merck-NM_145060_at SKA1 merck-NM_198436_s_at AURKA merck2-NM_001039535_a_at SKA1 merck2-NM_145060_a_at SKA1 merck-ENST00000333706_x_at BIRC5 merck-AK223428_a_at BIRC5 merck-NM_004219_x_at PTTG1 merck-NM_012310_at KIF4A GDPD2 merck-NM_001809_at CENPA merck2-ENST00000333706_s_at merck-NM_001276_at CHI3L1 merck-NM_018101_at CDCA8 merck-ENST00000360566_at RRM2 merck2-BC001651_at CDCA8 merck2-AF098158_at TPX2 merck-NM_012112_at TPX2 merck-NM_005733_at KIF20A CDC23 merck-U63743_a_at KIF2C merck2-AK123247_at MYH11 NDE1 merck2-ENST00000331944_s_at merck-NM_181802_at UBE2C merck2-NM_018410_at HJURP merck2-BT006759_at KIF2C merck2-M87338_at RFC2 merck-NM_152637_at METTL7B ITGA7 merck-NM_182513_at SPC24 merck-NM_018154_at ASF1B PRKACA merck2-AL519719_a_at BIRC5 merck2-BC007417_at POC1A merck-NM_021953_at FOXM1 merck-NM_016426_at GTSE1 TRMU merck-CR602926_s_at CCNB1 merck-NM_014791_at MELK merck-NM_006342_at TACC3 merck-NM_004701_at CCNB2 merck-NM_004217_at AURKB merck-NM_144569_s_at SPOCD1 merck2-NM_001168_at BIRC5 merck2-BC006325_at GTSE1 TRMU merck-NM_018131_at CEP55 merck-AY605064_at CLSPN merck-NM_004336_at BUB1 RGPD6 merck-NM_031299_at CDCA3 GNB3 merck2-AF043294_at BUB1 RGPD6 merck2-NM_014397_at NEK6 merck-NM_001255_s_at CDC20 merck2-ENST00000370966_a_at DEPDC1 OTUD7A merck-ENST00000243201_a_at HJURP merck-NM_003258_at TK1 merck-CR602847_a_at KIAA0101 merck-NM_006547_at IGF2BP3 AMOTL1 MALSU1 merck2-BC006325_x_at GTSE1 TRMU merck-BC075828_a_at GTSE1 merck-NM_014750_at DLGAP5 merck-NM_203394_at E2F7 merck-ENST00000308604_s_at LINC00152 MIR4435-1HG merck-AF469667_a_at MLF1IP merck-BI868409_a_at MKI67 merck-NM_016639_at TNFRSF12A CLDN9 merck-CR607300_a_at MKI67 merck-NM_001237_a_at CCNA2 EXOSC9 merck-NM_152515_at CKAP2L merck-AK055931_a_at SHCBP1 merck-NM_005192_at CDKN3 merck2-AK000490_a_at DEPDC1 merck-NM_012291_at ESPL1 PFDN5 merck-BC106033_s_at SMC4 merck2-BC034607_at ASPM merck-NM_152562_s_at CDCA2 merck-NM_004237_at TRIP13 merck2-AK026140_at merck-NM_001813_at CENPE merck2-BC005978_at KPNA2 merck2-NM_024745_at SHCBP1 merck-CR610123_a_at POC1A merck-NM_001790_at CDC25C merck2-Y00472_a_at SOD2 merck2-BC025232_at CDC6 merck2-NM_017779_at DEPDC1 merck-NM_004526_at MCM2 merck2-BC107750_at CDK1 RHOBTB1 merck-BX649059_at GAS2L3 merck-NM_005480_at TROAP merck-NM_007243_a_at NRM merck2-NM_031966_at CCNB1 merck-NM_001024466_s_at SOD2 merck2-BC005978_s_at KPNA2 merck-NM_080668_at CDCA5 merck-NM_004911_at PDIA4 merck-BC004202_a_at CHEK1 merck-NM_003504_at CDC45 merck2-BC098582_at KIF14 merck2-M36693_a_at SOD2 merck-NM_012145_a_at DTYMK merck-NM_017581_at CHRNA9 merck2-BM464374_at CENPE merck-NM_001845_at COL4A1 merck2-DQ890621_at CDC45

TABLE 2 100 immune signature genes probe Gene merck-NM_003151_a_at STAT4 merck2-AJ515553_at AMICA1 merck-NM_153206_s_at AMICA1 merck-NM_006682_s_at FGL2 CCDC146 merck-NM_000733_at CD3E merck-BC030533_s_at TRBC1 TRBV19 merck-NM_001767_at CD2 merck-BC014239_s_at PTPRC merck-NM_001040067_s_at TRBC2 TRBV3-1 TRBV5-4 TRBV6-5 TRBV7-2 merck-NM_002209_at ITGAL merck-NM_080612_at GAB3 merck2-ENST00000390420_at TRBV3-1 TRBV5-4 TRBV6-5 TRBV7-2 merck2-AA669142_at merck-NM_002104_at GZMK merck-NM_005546_at ITK CYFIP2 merck-NM_018384_at GIMAP5 GIMAP1-GIMAP5 merck2-ENST00000390409_at TRBC1 TRBV19 merck-NM_153236_at GIMAP7 merck2-ENST00000390420_s_at merck2-ENST00000390537_s_at merck-NM_003650_at CST7 merck-NM_001504_at CXCR3 merck-NM_000732_at CD3D merck-AI281804_at GPR174 merck-ENST00000382913_s_at TRAC TRAJ17 TRAV20 TRDV2 merck2-NM_198196_a_at CD96 merck-NM_001558_at IL10RA merck-NM_002832_at PTPN7 merck-NM_005335_at HCLS1 merck2-NM_001558_at IL10RA merck2-AL833681_at CD96 merck-NM_175900_s_at C16orf54 QPRT merck-AK021632_at ANKRD44 merck2-NM_175900_at C16orf54 QPRT merck-NM_003978_at PSTPIP1 merck-NM_032214_at SLA2 merck-NM_014207_at CD5 merck2-NM_005816_a_at CD96 merck2-NM_001114380_x_at ITGAL merck2-DB317311_at GIMAP1 merck-NM_001781_at CD69 merck-NM_030767_at AKNA merck-ENST00000318430_s_at TMC8 merck2-AW798052_at AKNA merck2-NM_002209_x_at ITGAL merck-NM_016388_at TRAT1 merck-NM_002298_s_at LCP1 merck-NM_007360_at KLRK1 KLRC4-KLRK1 merck-NM_024070_at PVRIG merck-NM_005816_at CD96 merck2-BM977026_at merck-NM_017424_at CECR1 merck-NM_032496_at ARHGAP9 merck-NM_130848_s_at C5orf20 merck2-NM_177405_a_at CECR1 merck-NM_001037631_at CTLA4 ICOS merck2-NM_145642_a_at APOL3 merck-BC017813_a_at FGL2 CCDC146 merck-AK025758_at NFATC2 merck2-NM_014349_a_at APOL3 merck2-NM_145640_a_at APOL3 merck-BE856897_s_at NFATC2 merck2-NM_030644_a_at APOL3 merck2-NM_145639_a_at APOL3 merck-ENST00000381961_at IL7R merck2-AA278761_at merck-NM_014716_at ACAP1 merck-NM_000206_at IL2RG merck2-NM_007360_at KLRK1 KLRC4-KLRK1 merck-ENST00000343625_s_at RASAL3 merck-BG271748_s_at GIMAP1 merck-NM_000734_at CD247 merck-NM_003387_at WIPF1 merck-NM_005541_at INPP5D merck2-NM_145641_a_at APOL3 merck-BX648371_at LINC00861 merck2-NM_017424_a_at CECR1 merck-NM_001838_at CCR7 merck-CR617832_a_at MS4A1 merck2-BX640915_at TIGIT merck-NM_006725_at CD6 merck-NM_198517_at TBC1D10C merck-BC028068_s_at JAK3 INSL3 merck2-NM_006120_at HLA-DMA BRD2 merck-NM_001079_at ZAP70 merck-AF402776_at MIR155HG merck-NM_014879_at P2RY14 merck-NM_052931_at SLAMF6 merck-NM_022141_at PARVG merck-NM_018460_at ARHGAP15 merck-NM_001025265_at CXorf65 merck-NM_024898_s_at DENND1C CRB3 merck-NM_001001895_at UBASH3A merck-ENST00000316577_s_at TESPA1 merck2-BC020657_at GIMAP4 merck-NM_004877_at GMFG merck-M21624_s_at TRDC merck2-BM678246_at CD37 merck-NM_018556_s_at SIRPG merck-NM_145641_s_at APOL3

The number of genes in each pathway was reduced to 10 genes.

Proliferation:

    • Probe IDs: merck-NM_012112_at, merck-NM_001809 at merck-U63743_a_at, merck-NM_004701_at, merck2-AF043294_at, merck-ENST00000243201_a_at, merck-NM_080668_at, merck-NM_004219_x_at merck-NM_018131_at merck-NM_145060_at
    • Gene symbols: TPX2, CENPA, KIF2C, CCNB2, BUB1, HJURP, CDCA5, PTTG1, CEP55, SKA1

Immune Signature:

    • Probe IDs: merck-NM_000732_at, merck-NM_001767_at, merck-NM_000733_at, merck-NM_005546_at, merck2-ENST00000390409_at, merck-NM_198517_at, merck-NM_014716_at, merck-NM_000734_at, merck-NM_052931_at, merck2-B1519527_at
    • Gene symbols: CD3D, CD2, CD3E, ITK, TRBC1, TBC1D10C, ACAP1, CD247, SLAMF6, IKZF1

The scores derived from these 10-genes correlated to the original scores at the level of 0.99 for both proliferation and immune score. The formula for calculating the prediction score is:


Breast Cancer Risk Score=0.404457 (−0.026432*ER)+(−0.001974*HER2)+(0.034656*Proliferation)+(−0.054045*immune)+(0.127414*stage)   (Formula 2).

This model predicts breast cancer patient outcome (overall survival) in 1249 primary breast cancer validation set. For example, at the threshold of 0.2, the odds ratio is 5.31 (95% CI: 3.57-7.88). The Fisher's Exact Test P-value is 9.8×10−20.

The validation patients can be further divided into good, medium and poor prognosis groups. FIG. 3 shows the Kaplan-Meier curves for patients with prediction score<0.2 (good prognosis), 0.2-0.35 (medium prognosis) and >0.35 (poor prognosis) respectively. The P-value based on Chi-square test is 0.

The risk of death increases linearly with the prediction score. Table 3 illustrates the death rate and bone metastasis rate vs. prediction scores.

TABLE 3 Death rate and bone metastasis rate verses prediction score Prediction Number of Number of Bone Mets score samples deaths Death rate Bone mets rate <0 110 1 0.009 0 0.000   0-0.1 252 12 0.048 0 0.000 0.1-0.2 300 21 0.070 7 0.023 0.2-0.3 278 40 0.144 7 0.025 0.3-0.4 166 36 0.217 14 0.084 >0.4 143 55 0.385 19 0.133

Example 2 Prognostic Model for Lung Cancer

This example describes a lung cancer prognosis model which uses gene expression profiling data and tumor stage. The model contains multiple gene expression signatures as components and the tumor stage. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

There are numerous studies of prognoses using gene expression alone, or histopathology/clinical data alone. Here we combine both to further improve the prognosis.

A total of 2,978 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model validated using the second half of samples. In the first half of samples, 1,456 samples had outcome data (live or death), and 1,339 patients had tumor stage measurement. In the second half of samples, 1,486 had outcome data, and 1,168 patients had stage measurement.

The model was built in the training set using a general linear model (from the R package) using the following equation:


Lung Cancer Risk Score=−0.54238+(−0.04826*imscore)+(0.04317*hscore)+(0.03468*ras)+(−0.01188*prg)+(0.09167*pscore)+(0.07474*stage)   (Formula 3),

where “imscore” is an immune score calculated from immune signature genes in Table 4, “hscore” is a hypoxia score from hypoxia signature genes in Table 5, “ras” is a score from ras signature genes in Table 6, “prg” is a score calculated from prognosis genes listed in Table 7, “pscore” is a proliferation score from the proliferation signature genes in Table 8, and the stage is the composite tumor stage. Scores for each signature was computed simply by averaging the log2 expression level of the genes in the signature.

TABLE 4 Immune signature genes probe Gene merck-NM_005356_at LCK merck-NM_006144_at GZMA merck-NM_014207_at CD5 merck-NM_005608_at PTPRCAP merck-NM_007181_at MAP4K1 merck-NM_002738_at PRKCB merck-Y00638_s_at PTPRC merck-BC014239_s_at PTPRC merck-NM_130446_at KLHL6 merck-NM_005546_at ITK CYFIP2 merck-NM_006257_at PRKCQ merck-NM_002104_at GZMK merck-NM_001504_at CXCR3 merck-NM_001001895_at UBASH3A merck-NM_002832_at PTPN7 merck-NM_018460_at ARHGAP15 merck-NM_001838_at CCR7 merck-NM_002209_at ITGAL merck-NM_006725_at CD6 merck-BC028068_s_at JAK3 INSL3 merck-NM_001079_at ZAP70 merck-NM_005541_at INPP5D merck-ENST00000318430_s_at TMC8 merck-NM_006564_at CXCR6 merck-NM_007237_s_at SP140 merck-NM_178129_at P2RY8 merck-NM_000647_s_at CCR2 merck-BU428565_s_at P2RY8 merck-NM_002351_s_at SH2D1A merck-NM_001040033_at CD53 merck-NM_005816_at CD96 merck-NM_198517_at TBC1D10C merck-NM_000733_at CD3E merck-NM_002163_at IRF8 merck-NM_000655_at SELL merck-NM_003037_at SLAMF1 merck-NM_003151_a_at STAT4 merck-NM_001007231_s_at ARHGAP25 merck-NM_018326_at GIMAP4 merck-NM_000377_at WAS merck-NM_001558_at IL10RA merck-NM_002985_at CCL5 merck-DT807100_at CD3D CD3G merck-NM_001465_at FYB merck-BP339517_a_at FYB merck-NM_030767_at AKNA merck-NM_005565_at LCP2 merck-NM_001040031_at CD37 merck-NM_002872_at RAC2 merck-NM_019604_at CRTAM merck-NM_005263_at GFI1 merck-NM_001037631_at CTLA4 ICOS merck-NM_016388_at TRAT1 merck-NM_014450_at SIT1 RMRP merck-NM_000732_at CD3D merck-NM_000073_at CD3G merck-NM_007360_at KLRK1 KLRC4-KLRK1 merck-NM_013351_at TBX21 merck-NM_032214_at SLA2 merck-NM_000639_at FASLG merck-NM_001242_at CD27 merck-ENST00000381961_at IL7R merck-NM_153206_s_at AMICA1 merck-NM_001025598_at ARHGAP30 USF1 merck-NM_001768_at CD8A merck-NM_003978_at PSTPIP1 merck-NM_014716_at ACAP1 merck-AK128740_s_at IL16 merck-NM_006060_a_at IKZF1 merck-BC075820_at IKZF1 merck-NM_016293_at BIN2 merck-NM_012092_at ICOS merck-NM_005442_at EOMES LOC100996624 merck-NM_007074_at CORO1A merck-NM_000206_at IL2RG merck-NM_005041_at PRF1 merck-NM_024898_s_at DENND1C CRB3 merck-NM_173799_at TIGIT merck-NM_001767_at CD2 merck-NM_002348_at LY9 merck-X60502_s_at SPN QPRT merck-NM_153236_at GIMAP7 merck-NM_005601_at NKG7 merck-NM_032496_at ARHGAP9 merck-NM_004877_at GMFG merck-NM_021181_at SLAMF7 merck-NM_018384_at GIMAP5 GIMAP1-GIMAP5 merck-NM_181780_at BTLA merck-NM_001017373_at SAMD3 merck-NM_000734_at CD247 merck-NM_003650_at CST7 merck-NM_172101_at CD8B merck-NM_001803_at CD52 merck-NM_001778_at CD48 merck-NM_001025265_at CXorf65 merck-NM_198929_at PYHIN1 merck-ENST00000379833_at GVINP1 merck-NM_052931_at SLAMF6 merck-NM_001024667_s_at FCRL3 merck-NM_002258_at KLRB1 merck-NM_018556_s_at SIRPG merck-AK090431_s_at NLRC3 merck-NM_018990_at SASH3 XPNPEP2 merck-NM_175900_s_at C16orf54 QPRT merck-ENST00000316577_s_at TESPA1 merck-NM_024070_at PVRIG merck-AY190088_s_at merck-NM_001040067_s_at TRBC2 TRBV3-1 TRBV5-4 TRBV6-5 TRBV7-2 merck-NM_130848_s_at C5orf20 merck-ENST00000381153_at C11orf21 merck-ENST00000382913_s_at TRAC TRAJ17 TRAV20 TRDV2 merck-BC030533_s_at TRBC1 TRBV19 merck-ENST00000244032_a_at ZNF831 merck-ENST00000371030_at ZNF831 merck-ENST00000343625_s_at RASAL3 merck-AF143887_at merck-AK128436_at IKZF3 merck-AI281804_at GPR174 merck-AF086367_at merck-CR598049_at LINC00426 merck-BM700951_at KLRK1 KLRC4-KLRK1 merck-BX648371_at LINC00861 merck-BC070382_at merck2-AW798052_at AKNA merck2-BX640915_at TIGIT merck2-BM678246_at CD37 merck2-NM_025228_at TRAF3IP3 merck2-XM_033379_at WDFY4 merck2-AJ515553_at AMICA1 merck2-BP262340_at IL16 merck2-AK225623_at DENND1C CRB3 merck2-AL833681_at CD96 merck2-BF111803_at ARHGAP15 merck2-BX406128_at CD3G merck2-NM_153701_at merck2-BC020657_at GIMAP4 merck2-AY185344_at PYHIN1 merck2-DR159064_at EOMES LOC100996624 merck2-ENST00000390420_at TRBV3-1 TRBV5-4 TRBV6-5 TRBV7-2 merck2-ENST00000390420_s_at merck2-NM_001010923_at THEMIS merck2-ENST00000390409_at TRBC1 TRBV19 merck2-AX721088_at merck2-ENST00000390393_at TRBV19 merck2-AW341086_at merck2-AA278761_at merck2-AA278761_x_at merck2-ENST00000390394_s_at merck2-AA669142_at merck2-AW007991_at PTPRC merck2-BG743900_at PRKCB merck2-X06318_at PRKCB merck2-BI519527_at IKZF1 merck2-ENST00000390537_s_at merck2-AY292266_x_at merck2-NM_005816_a_at CD96 merck2-NM_198196_a_at CD96 merck2-NM_001114380_x_at ITGAL merck2-NM_007237_a_at SP140 merck2-NM_007237_at SP140 merck2-NM_052931_at SLAMF6 merck2-NM_001558_at IL10RA merck2-NM_007360_at KLRK1 KLRC4-KLRK1 merck2-NM_002209_x_at ITGAL merck2-NM_175900_at C16orf54 QPRT

TABLE 5 Hypoxia signature genes probe Gene merck-NM_002627_at PFKP PITRM1 merck-NM_000302_at PLOD1 merck-NM_001216_at CA9 RMRP merck-ENST00000377093_at KIF1B merck-BC004202_a_at CHEK1 merck-NM_030949_at PPP1R14C merck-CR593119_a_at CLIC4 merck-NM_001255_s_at CDC20 merck-BG679113_s_at KRT6A KRT6B KRT6C merck-NM_002421_at MMP1 merck-BQ217236_a_at SERPINB5 merck-NM_001793_at CDH3 merck-NM_001238_at CCNE1 merck-BU597348_s_at SYNCRIP merck-NM_006516_at SLC2A1 merck-BX648425_a_at DSC2 merck-X15014_a_at RALA merck-NM_018685_at ANLN merck-CR614206_a_at ERO1L merck-NM_001124_at ADM merck-NM_015440_at MTHFD1L merck-ENST00000367307_a_at MTHFD1L merck-NM_058179_at PSAT1 merck-NM_031415_s_at GSDMC merck-NM_005557_x_at KRT16 merck-NM_053016_at PALM2 PALM2-AKAP2 merck-CR602579_a_at CTPS1 merck-NM_001428_s_at ENO1 merck-ENST00000305850_at CENPN CMC2 merck-NM_005978_at S100A2 merck-NM_018643_at TREM1 merck-NM_006505_at PVR merck-NM_080655_s_at MSANTD3 merck-NM_001012507_at CENPW merck-ENST00000258005_a_at NHSL1 merck-AK129763_at LINC00673 merck-XM_927868_s_at PGK1 merck-XM_928117_x_at FAM106B merck-AL359337_at ADM merck-AA148856_s_at SYNCRIP merck2-AI989728_at SERPINB5 merck2-DQ892208_at CA9 RMRP merck2-AK022036_at WWTR1 merck2-AA677426_at merck2-AA677426_s_at merck2-BC004856_at NCS1 merck2-BG252150_at PFKP merck2-BC007633_at AGO2 merck2-BG400371_at merck2-DQ891441_at merck2-NM_017522_AS_at LRP8 merck2-AF039652_at RNASEH1 merck2-AV714642_at ANLN merck2-AB030656_at CORO1C merck2-NM_000291_at PGK1 merck2-NM_005554_at KRT6A merck2-BC002829_at S100A2 merck2-BU681245_at merck2-AK225899_a_at CTPS1 merck2-BC062635_a_at XPO5 merck2-AF257659_a_at CALU merck2-CA308717_at merck2-X56807_at DSC2 merck2-CR936650_at ANLN merck2-AY423725_a_at PGK1 merck2-BC103752_a_at PGK1

TABLE 6 Ras signature genes probe Gene merck-NM_002205_at ITGA5 merck-NM_000376_at VDR merck-NM_002203_at ITGA2 merck-NM_002658_at PLAU merck-CD014069_s_at TNFRSF1A merck-NM_004419_at DUSP5 merck-NM_021199_s_at SQRDL merck-NM_016639_at TNFRSF12A CLDN9 merck-NM_002068_at GNA15 merck-NM_005562_at LAMC2 merck-BG677853_a_at LAMC2 merck-BM980789_s_at LAMC2 merck-ENST00000265539_s_at FOSL2 merck-NM_013451_at MYOF merck-ENST00000371489_s_at MYOF merck-NM_003670_at BHLHE40 merck-NM_000577_s_at IL1RN merck-NM_000228_at LAMB3 merck-NM_003897_a_at IER3 LINC00243 merck-NM_003955_at SOCS3 merck-NM_001002857_at ANXA2 merck-NM_080388_at S100A16 merck-NM_022162_at NOD2 merck-NM_003461_at ZYX merck-NM_002966_at S100A10 merck-NM_004240_at TRIP10 merck-NM_005194_at CEBPB merck-NM_005620_at S100A11 merck-NM_002090_at CXCL3 merck-NM_000418_at IL4R merck-NM_001005377_s_at PLAUR merck-NM_001005376_at PLAUR merck-NM_001511_at CXCL1 merck-BC053563_s_at MIR21 merck-ENST00000333244_at AHNAK2 merck2-AI701192_at LAMC2 merck2-AI701192_x_at LAMC2 merck2-AI858819_at merck2-AK075141_at RNF149 merck2-AK092006_s_at merck2-CA445253_at MYOF merck2-BT009912_at merck2-BT009912_x_at merck2-NM_000700_at ANXA1 merck2-BC001405_at UPP1 merck2-NM_001005377_at PLAUR merck2-M62898_x_at ANXA2 merck2-BG680883_at merck2-BC082238_at BHLHE40 merck2-BG675923_x_at merck2-BM543893_x_at PLAUR merck2-X74039_at PLAUR

TABLE 7 Prognosis signature genes probe Gene merck-CN269476_a_at PCDP1 merck-NM_002126_at HLF merck-NM_031911_a_at C1QTNF7 merck2-BX647781_at C1QTNF7 merck-NM_000901_at NR3C2 merck-NM_021117_at CRY2 merck-BU681386_at SCN7A merck2-AI949138_at PCDP1 merck-AJ315514_a_at NR3C2 merck-NM_153267_at MAMDC2 merck-NM_007037_at ADAMTS8 merck2-BM684168_at merck-NM_006030_at CACNA2D2 merck-NM_001029996_at PCDP1 merck-NM_033053_s_at DMRTC1 DMRTC1B merck2-NM_001080851_s_at merck2-BC128418_at CBX7 merck-AK057720_s_at OBFC1 merck-NM_002976_at SCN7A merck-AI027436_at merck-AL832580_at RNF180 merck-NM_004962_at GDF10 merck-AK124663_a_at WDFY3-AS2 merck-AF329839_a_at C1QTNF7 merck2-CB999963_at RNF180 merck-NM_175709_at CBX7 merck-NM_007106_at UBL3 merck-AA129758_a_at EIF4E3 merck-AK023631_at merck2-BC036093_at HLF merck2-BM976317_at ANKDD1B merck-BC038509_a_at RCAN2 merck2-NM_020139_at BDH2 merck-NM_004469_at FIGF PIR-FIGF merck-BQ709647_a_at HLF merck-BG678236_at SAR1B merck-NM_152606_at ZNF540 merck-NM_007168_at ABCA8 merck2-NM_020139_a_at BDH2 merck2-AL832100_at ZNF540 merck-AK090989_at merck-NM_030569_at ITIH5 merck-NM_014774_at EFCAB14 merck-NM_183075_at CYP2U1 merck-NM_020899_s_at ZBTB4 merck-BC095414_a_at BDH2 merck-NM_032411_at C2orf40 merck2-H45244_at merck-NM_006856_at ATF7 LOC100652999 merck-NM_018488_at TBX4 merck-NM_018010_at IFT57 merck-NM_021965_s_at PGM5 merck2-BC062365_at SLIT3 merck-NM_172193_at KLHDC1 merck-NM_005181_at CA3 merck-CX782760_at TAPT1 merck-DB366031_s_at CREBRF merck-NM_199454_at PRDM16 merck2-AI478811_at EMCN merck-ENST00000374232_at SNX30 merck-NM_001008710_s_at RBPMS merck-NM_152459_at C16orf89 SEC14L5 merck-AK075495_at NDFIP1 merck2-CN308012_at EFCAB14 merck-NM_021977_at SLC22A3 merck-BX537534_at BTBD9 merck-NM_001174_s_at ARHGAP6 merck-AY312852_s_at GTF2IRD2 GTF2IRD2B GTF2I merck-NM_003206_a_at TCF21 merck2-NM_001018108_at SERF2 merck-NM_014880_at CD302 LY75-CD302 merck-NM_030923_s_at TMEM163 merck-AL133118_at EMCN merck2-BG674122_a_at HLF merck-NM_003099_at SNX1 CSNK1G1 merck-AL161983_at EIF4E3 merck2-NM_173537_s_at merck-AK130274_at merck-BC073920_at LOC100652999 merck-NM_004614_s_at TK2 merck-NM_198901_at SRI merck2-NM_024768_at EFCC1 merck2-CR598366_at merck-NM_014701_at SECISBP2L merck-ENST00000382101_a_at DLC1 merck-NM_015328_at AHCYL2 merck-BX106890_a_at ITGA8 LOC101928678 merck-BC023330_at LINC00849 merck-NM_014232_at VAMP2 merck-BC050653_a_at NICN1 AMT merck-AK096254_at merck-ENST00000283296_a_at GPR116 LOC101926962 merck2-BX115850_at IFT57 merck-NM_032866_at CGNL1 merck-NM_174934_at SCN4B merck-NM_024513_s_at FYCO1 merck2-NM_001003795_s_at merck-NM_021902_s_at FXYD1 merck-NM_152913_at TMEM130 merck-BC030082_at SORBS2

TABLE 8 Proliferation signature genes probe Gene merck-NM_003318_at TTK merck-NM_014791_at MELK merck-NM_001786_a_at CDK1 RHOBTB1 merck-NM_001790_at CDC25C merck-NM_014176_at UBE2T merck-BF511624_s_at BUB1B merck-NM_005030_at PLK1 merck-NM_181802_at UBE2C merck-NM_004217_at AURKB merck-NM_201567_at CDC25A merck-NM_198436_s_at AURKA merck-NM_001255_s_at CDC20 merck-NM_003579_at RAD54L merck-NM_004336_at BUB1 RGPD6 merck-NM_031299_at CDCA3 GNB3 merck-NM_004237_at TRIP13 merck-BC001459_s_at RAD51 merck-NM_012484_at HMMR merck-AB042719_a_at MCM10 merck-NM_018518_at MCM10 merck-NM_012291_at ESPL1 PFDN5 merck-NM_014750_at DLGAP5 merck-NM_199413_at PRC1 merck-NM_130398_at EXO1 merck-NM_199420_s_at POLQ merck-NM_005733_at KIF20A CDC23 merck-NM_004856_at KIF23 merck-NM_004701_at CCNB2 merck-NM_014321_at ORC6 merck-NM_002466_at MYBL2 merck-NM_030919_at FAM83D merck-NM_003504_at CDC45 merck-BC075828_a_at GTSE1 merck-NM_016426_at GTSE1 TRMU merck-NM_001012409_at SGOL1 merck-NM_018136_s_at ASPM merck-NM_018685_at ANLN merck-NM_012112_at TPX2 merck-NM_018101_at CDCA8 merck-NM_001237_a_at CCNA2 EXOSC9 merck-NM_018454_at NUSAP1 merck-NM_001211_at BUB1B merck-U63743_a_at KIF2C merck-CR596700_a_at RRM2 merck-NM_012310_at KIF4A GDPD2 merck-NM_013277_a_at RACGAP1 merck-NM_018154_at ASF1B PRKACA merck-BC024211_a_at NCAPH merck-NM_152515_at CKAP2L merck-NM_018131_at CEP55 merck-NM_002417_at MKI67 merck-CR607300_a_at MKI67 merck-BI868409_a_at MKI67 merck-NM_001813_at CENPE merck-CR602926_s_at CCNB1 merck-NM_001809_at CENPA merck-NM_080668_at CDCA5 merck-AK223428_a_at BIRC5 merck-NM_005480_at TROAP merck-NM_021953_at FOXM1 merck-NM_144508_at CASC5 merck-NM_019013_at FAM64A PITPNM3 merck-hCT1776373.2_s_at DEPDC1 OTUD7A merck-NM_004091_at E2F2 merck-NM_004219_x_at PTTG1 merck-NM_002263_a_at KIFC1 merck-AF331796_a_at NCAPG merck-NM_145060_at SKA1 merck-BC048988_a_at SKA3 merck-NM_152259_s_at TICRR KIF7 merck-ENST00000243201_a_at HJURP merck-ENST00000333706_x_at BIRC5 merck-ENST00000335534_s_at KIF18B merck-AY605064_at CLSPN merck2-AK097710_at CDC25C merck2-AF043294_at BUB1 RGPD6 merck2-AU132185_at MKI67 merck2-BC098582_at KIF14 merck2-BT006759_at KIF2C merck2-BC006325_at GTSE1 TRMU merck2-BC006325_x_at GTSE1 TRMU merck2-AL832036_at CKAP2L merck2-DQ890621_at CDC45 merck2-NM_005196_at CENPF merck2-AV714642_at ANLN merck2-BC034607_at ASPM merck2-BC001651_at CDCA8 merck2-AF098158_at TPX2 merck2-NM_001168_at BIRC5 merck2-AK023483_at NUSAP1 merck2-NM_145061_at SKA3 merck2-NM_018410_at HJURP merck2-AL517462_s_at merck2-ENST00000333706_s_at merck2-BX648516_at SGOL1 merck2-AK000490_a_at DEPDC1 merck2-ENST00000370966_a_at DEPDC1 OTUD7A merck2-AB046790_at CASC5 merck2-CR936650_at ANLN merck2-AL519719_a_at BIRC5 merck2-NM_145060_a_at SKA1 merck2-NM_001039535_a_at SKA1

The performance of this model was evaluated in reserved validation set of 1,486 samples. FIG. 4 shows the predicted death rate vs. the actual average (running average of 200 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 9.

TABLE 9 Average death rate versus prediction score. Prediction Number of Number of score samples deaths Rate <0.3 151 25 0.165562914 0.3-0.4 132 25 0.189393939 0.4-0.5 171 68 0.397660819 0.5-0.6 207 94 0.45410628 0.6-0.7 203 118 0.581280788 0.7-0.8 144 82 0.569444444 >0.8 160 122 0.7625

Using a threshold of 0.4, the odds ratio for overall survival was 5.62 (95% CI: 4.03-7.85), Fisher's Exact Test p-value=2.9×10−29.

Patients can be further divided into good (risk score<0.4), medium (score 0.4-0.7) and poor (score>0.7) prognosis groups. FIG. 5 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 128 (P=0).

The number of genes in each pathway was reduced to 10 genes.

Immune Signature:

    • Probe IDs: merck-NM_001767_at, merck2-NM_002209_x_at, merck2-BI519527_at, merck-NM_000732_at, merck2-ENST00000390409_at, merck-NM_014716_at, merck-NM_000733_at, merck-NM_198517_at, merck-NM_000734_at, merck2-NM_052931_at
    • Gene symbols: CD2, ITGAL, IKZF1, CD3D, TRBC1, ACAP1, CD3E, TBC1D10C, CD247, SLAMF6

Hypoxia:

    • Probe IDs: merck-NM_006516_at, merck2-BC002829_at, merck-NM_005557_x_at, merck2-NM_005554_at, merck-BX641095_a_at, merck-NM_024009_at, merck-NM_006142_at, merck-NM_033386_s_at, merck-NM_020183_s_at, merck-NM_000094_at
    • Gene symbols: SLC2A1, S100A2, KRT16, KRT6A, CD109, GJB3, SFN, MICALL1, ARNTL2, COL7A1

Ras Signature:

    • Probe IDs: merck-NM_005620_at, merck2-AI701192_at, merck2-M62898_x_at, merck-NM_002658_at, merck2-X74039_at, merck-NM_080388_at, merck-NM_000418_at, merck-NM_002068_at, merck-NM_013451_at, merck-NM_000228_at
    • Gene symbols: S100A11, LAMC2, ANXA2, PLAU, PLAUR, S100A16, IL4R, GNA15, MYOF, LAMB3

Prognosis:

    • Probe IDs: merck-NM_002126_at, merck-BU681386_at, merck-NM_000901_at, merck2-AI949138_at, merck-NM_007168_at, merck2-AI478811_at, merck-NM_018010_at, merck-BC095414_a_at, merck-NM_153267_at, merck-ENST00000378076_at
    • Gene symbols: HLF, SCN7A, NR3C2, PCDP1, ABCA8, EMCN, IFT57, BDH2, MAMDC2, ITGA8

Proliferation:

    • Probe IDs: merck-NM_012112 at merck-NM_001809_at merck-U63743_a_at merck-NM_004701_at merck-NM_080668_at merck-ENST00000243201_a_at merck-NM_012310_at merck-ENST00000333706_x_at merck-NM_014750_at merck-NM_145060_at
    • Gene symbols: TPX2, CENPA, KIF2C, CCNB2, CDCA5, HJURP, KIF4A, BIRC5, DLGAP5, SKA1

The scores derived from these 10-genes correlated to the original scores at the level of 0.99 for both proliferation and immune scores, 0.98 for ras signature, 0.97 for the prognosis signature and 0.92 for the hypoxia signature.

The ras signature was marginally predictive in the original model, and is not significant after the number of genes was reduced for all these pathways. Hence it was excluded from the model. The formula for the updated model (based on small number of genes) is:


Lung Cancer Risk Score=−0.2853866+(−0.0328615*imscore)+(0.0269496*hscore)+(−0.0006368*prg)+(0.0928468*pscore)+(0.0757314*stage)   (Formula 4).

Note, the exact coefficients change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

FIG. 6 shows the predicted death rate vs. the actual average (running average of 200 samples as ranked by the prediction score) death rate for this updated model. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 10.

TABLE 10 Average death rate versus prediction score. Prediction Number of Number of score samples deaths Rate <0.3 141 22 0.156028369 0.3-0.4 135 29 0.214814815 0.4-0.5 166 60 0.361445783 0.5-0.6 220 99 0.45 0.6-0.7 201 116 0.577114428 0.7-0.8 140 81 0.578571429 >0.8 165 127 0.76969697

Using a threshold of 0.4, the odds ratio for overall survival was 5.21 (95% CI: 3.74-7.26), Fisher's Exact Test p-value=7.3×10−27.

Patients can be further divided into good (risk score<0.4), medium (score 0.4-0.7) and poor (score>0.7) prognosis groups. FIG. 7 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 123 (P=0).

This multicomponent model included both microarray measurement and tumor stage. Each of the components is significant in the model according to the AVOVA analysis in the training set (Table 11).

TABLE 11 ANOVA test of fit model in the training set. Df Sum Sq Mean Sq F value Pr(>F) imscore_f[mke1] 1 5.123 5.1230 25.269 5.664e−07 *** hscore_f[mke1] 1 19.755 19.7553 97.444  <2.2e−16 *** prg1_f[mke1] 1 11.888 11.8880 58.638 3.623e−14 *** pscore_f[mke1] 1 11.084 11.0838 54.671 2.509e−13 *** stage[mke1] 1 8.959 8.9592 44.192 4.330e−11 *** Residuals 1333 270.247 0.2027

When microarray components (gene sets) were grouped together using the coefficients from the model, and applied to the validation set, the microarray part of the model was independently predictive of the patient outcome (FIG. 8). The F-static was 142.7 on 1 and 1166 degrees of freedom, P<2×10−16. The tumor stage was also a strong prognostic factor (F-static 103.9 on 1 and 1166 degrees of freedom P<2×10−16).

Example 3 Prognostic Model for Colon Cancer

This example describes a colon cancer prognosis model that uses gene expression profiling data and tumor stage. The model contains multiple gene expression signatures as components and the tumor stage. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

There are numerous studies of prognoses using gene expression alone, or histopathology/clinical data alone. Here both are combined to further improve the prognosis.

A total of 2,233 samples were profiled by Affymetrix® expression arrays, among them, 2,203 samples had outcome data (survival vs. death). A composite model was built using the first half of samples and the model validated using the second half of samples. In the first half of samples, 1,091 samples had outcome data (live or death), and 1,076 patients had tumor stage measurement. In the second half of samples, 1,112 had outcome data, and 1,057 patients had stage measurement.

A colon cancer risk model was built in the training set using a general linear model (from the R package) using the following equation:


Colon Cancer Risk Score=−1.109036+(−0.003155 *imscore)+(0.056980*hscore)+(−0.059340*emtscorel)+(−0.040061*emtscore2)+(−0.013334*prg1)+(0.285552*prg2)+(−0.015176*prg3)+(0.084259*stage)   (Formula 5),

where “imscore” is an immune score calculated from the immune signature gene in Table 11, “hscore” is a hypoxia score from hypoxia signature genes in Table 13, “emtscorel” is a score from the VIM correlated genes in Table 14, “emtscore2” is a score from the CDH1 correlated genes in Table 15, “prg1” is a score from prognosis genes in Table 16, “prg2” is a score from prognosis genes in Table 17, “prg3” is a score from prognosis genes in Table 18, and “stage” is the composite tumor stage. Scores from the signatures genes were computed simply by averaging the log2 expression level of the genes in the signature.

The performance of this model was evaluated using the reserved validation set of 1,057 samples. FIG. 9 shows the predicted death rate vs. the actual average (running average of 200 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 19.

TABLE 19 Average death rate versus prediction score Prediction Number of Number of score samples deaths Rate <0.2 179 20 0.111731844 0.2-0.3 178 39 0.219101124 0.3-0.4 194 45 0.231958763 0.4-0.5 220 90 0.409090909 >0.5 286 149 0.520979021

Using a threshold of 0.48, the odds ratio for overall survival was 3.47 (95% CI: 2.63-4.59), Fisher's Exact Test p-value=1.5×10−17.

Patients can be further divided into good (risk score<0.2), medium (score 0.2-0.5) and poor (score>0.5) prognosis groups. FIG. 10 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 52.6 (P=3.86×10−12). If the model is applied to the stage 1, 2, 3 patients (excluding stage 4) in the validation set, the Chi-square is 30.5 on 2 degrees of freedom (P=2.3×10−7, patients in 3 groups, Risk score<0.2, 0.2-0.5 and >0.5). The model is still predictive even if applied to stage 1 & 2 patients in the validation set. The Chi-square is 20.5 on 2 degrees of freedom (P=3.6×10−5, patients in 3 groups: Risk score<0.2, 0.2-0.4 and >0.4).

The number of genes in each pathway was reduced to 10 genes or less.

Immune signature:

    • Probe IDs: merck2-BI519527_at, merck2-NM_002209_x_at, merck-NM_001767_at, merck-NM_005546_at, merck-NM_007181_at, merck-NM_000733_at, merck-NM_198517_at, merck-NM_001040067_s_at, merck-NM_000734_at, merck-NM_000732 at
    • Gene symbols: IKZF1, ITGAL, CD2, ITK, MAP4K1, CD3E, TBC1D10C, TRBC2, CD247, CD3D

Hypoxia:

    • Probe IDs: merck-NM_006516_at, merck-X15014_a_at, merck-CR614206_a_at, merck-NM_018685_at, merck-NM_005978_at, merck2-AK223027_at, merck-NM_001255_s_at, merck-BG677853_a_at, merck2-X74039_at, merck2-NM001042422_at
    • Gene symbols: SLC2A1, RALA, ERO1L, ANLN, S100A2, PHLDA2, CDC20, LAMC2, PLAUR, SLC16A3

VIM Correlated Signature:

    • Probe IDs: merck2-AB266387_s_at,merck2-BQ632060_x_at, merck-ENST00000311127_a_at, merck2-NM_015463_at, merck-NM_006868_at, merck-BU625463_s_at, merck-AK091332_at, merck-NM_012219_s_at, merck-NM_144601_at, merck-NM_003255_s_at
    • Gene symbols: CCDC80, VIM, HEG1, CNRIP1, RAB31, EFEMP2, GNB4, MRAS, CMTM3, TIMP2

CDH1 Correlated Signature:

    • Probe IDs: merck-NM_004433_a_at, merck2-NM_001307_at, merck2-NM_001305_at, merck-NM_004360_at, merck-NM_020387_at, merck2-CK818800_at, merck-BC069241_a_at, merck2-NM_001982_at, merck-NM_005498_at, merck-ENST00000378957_a_at
    • Gene symbols: ELF3, CLDN7, CLDN4, CDH1, RAB25, ESRP1, ESRP2, ERBB3, AP1M2, EPCAM

Prognosis Component 1:

    • Probe IDs: merck-NM_002126_at, merck-BU681386_at, merck-NM_000901_at, merck2-AI949138_at, merck-NM_007168_at, merck2-AI478811_at, merck-NM_018010_at, merck-BC095414_a_at, merck-NM_153267_at, merck-ENST00000378076_at
    • Gene symbols: MZB1, OR6C4 IGKV3-11 IGKV3D-11 IGKV3D-20 RHNO1, TNFRSF17, IGKC IGKV1D-39 IGKV1-39, IGHA1 IGHG1 IGH, IGLC1, IGKC IGKV1-16 IGKV1D-16, IGLV6-57, IGLV1-40 IGLV5-39, IGJ

Prognosis Component 2:

    • Probe IDs: merck2-DQ892544_at, merck2-S42303_at, merck2-NM_133376_a_at, merck-BC010860_a_at, merck-AK125700_a_at, merck2-AL572880_at, merck2-EF043567_at, merck2-AI765059_at, merck2-CB115148_at, merck-NM_003254_at
    • Gene symbols: SPP1, CDH2, ITGB1, SERPINE1, PLOD2, COL4A1, NTM, MPRIP, PLIN2, TIMP1

The scores derived from these 10-genes correlated to the original scores at the level of 0.99 for both VIM and CDH1 correlated signature scores, and 0.98 for immune signature, 0.90 for the hypoxia signature, 0.99 for the prognosis component 1, and 0.90 for prognosis component 2.

Prognosis component 3 was marginally prognostic in the original model, and was not significant after the signatures reduced to 10 genes, hence was excluded from further models. The formula for the updated model (based on small number of genes) is:


Colon Cancer Risk Score=0.109098+(−0.029915*imscore)+(0.062785*hscore)+(−0.050770*emtscorel)+(−0.042210*emtscore2)+(−0.007858*prgl)+(0.099507*prg2)+(0.088208*stage)   (Formula 6).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

FIG. 11 shows the predicted death rate vs. the actual average (running average of 200 samples as ranked by the prediction score) death rate for this updated model. As shown in the figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 20.

TABLE 20 Average death rate versus prediction score. Prediction Number of Number of Score Samples Deaths Rate <0.2 115 13 0.113043478 0.2-0.3 148 24 0.162162162 0.3-0.4 233 59 0.253218884 0.4-0.5 232 82 0.353448276 0.5-0.6 175 83 0.474285714 >0.6 154 82 0.532467532

Using a threshold of 0.48, the odds ratio for overall survival was 3.03 (95% CI: 2.31-3.96), Fisher's Exact Test p-value=9.0×10−16.

Patients can be further divided into good (risk score<0.25), medium (score 0.25-0.5) and poor (score>0.5) prognosis groups. FIG. 12 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 57.2 (P=3.7×10−13).

This multicomponent model included both microarray measurement and tumor stage. Each of the components were significant in the model according to the AVOVA analysis in the training set (Table 21).

TABLE 21 ANOVA test of fit model in the training set. Df Sum Sq Mean Sq F value Pr(>F) imscore_f[mke1] 1 4.070 4.0698 18.6763 1.694e−05 *** hscore_f[mke1] 1 3.738 3.7384 17.1555 3.716e−05 *** emtscore1_f[mke1] 1 4.272 4.2722 19.6051 1.050e−05 *** emtscore2_f[mke1] 1 3.441 3.4413 15.7923 7.544e−05 *** prg1_f[mke1] 1 0.870 0.8705 3.9946 0.0459 * prg2_f[mke1] 1 7.949 7.9490 36.4783 2.128e−09 *** stage[mke1] 1 8.694 8.6937 39.8956 3.924e−10 *** Residuals 1068 232.730 0.2179

When microarray components (gene sets) were grouped together using the coefficients from the model, and applied to the validation set, the microarray part of the model was independently predictive of the patient outcome (FIG. 13). The F-static is 47.72 on 1 and 1055 degrees of freedom, P=8.5×10−12. The strongest prognostic factor was tumor stage (F-static 84.7 on 1 and 1055 degrees of freedom, P<2×10−16).

TABLE 12 Immune signature genes probe Gene merck-NM_005356_at LCK merck-NM_006144_at GZMA merck-NM_014207_at CD5 merck-NM_005608_at PTPRCAP merck-NM_007181_at MAP4K1 merck-NM_002738_at PRKCB merck-Y00638_s_at PTPRC merck-BC014239_s_at PTPRC merck-NM_130446_at KLHL6 merck-NM_005546_at ITK CYFIP2 merck-NM_006257_at PRKCQ merck-NM_002104_at GZMK merck-NM_001504_at CXCR3 merck-NM_001001895_at UBASH3A merck-NM_002832_at PTPN7 merck-NM_018460_at ARHGAP15 merck-NM_001838_at CCR7 merck-NM_002209_at ITGAL merck-NM_006725_at CD6 merck-BC028068_s_at JAK3 INSL3 merck-NM_001079_at ZAP70 merck-NM_005541_at INPP5D merck-ENST00000318430_s_at TMC8 merck-NM_006564_at CXCR6 merck-NM_007237_s_at SP140 merck-NM_178129_at P2RY8 merck-NM_000647_s_at CCR2 merck-BU428565_s_at P2RY8 merck-NM_002351_s_at SH2D1A merck-NM_001040033_at CD53 merck-NM_005816_at CD96 merck-NM_198517_at TBC1D10C merck-NM_000733_at CD3E merck-NM_002163_at IRF8 merck-NM_000655_at SELL merck-NM_003037_at SLAMF1 merck-NM_003151_a_at STAT4 merck-NM_001007231_s_at ARHGAP25 merck-NM_018326_at GIMAP4 merck-NM_000377_at WAS merck-NM_001558_at IL10RA merck-NM_002985_at CCL5 merck-DT807100_at CD3D CD3G merck-NM_001465_at FYB merck-BP339517_a_at FYB merck-NM_030767_at AKNA merck-NM_005565_at LCP2 merck-NM_001040031_at CD37 merck-NM_002872_at RAC2 merck-NM_019604_at CRTAM merck-NM_005263_at GFI1 merck-NM_001037631_at CTLA4 ICOS merck-NM_016388_at TRAT1 merck-NM_014450_at SIT1 RMRP merck-NM_000732_at CD3D merck-NM_000073_at CD3G merck-NM_007360_at KLRK1 KLRC4-KLRK1 merck-NM_013351_at TBX21 merck-NM_032214_at SLA2 merck-NM_000639_at FASLG merck-NM_001242_at CD27 merck-ENST00000381961_at IL7R merck-NM_153206_s_at AMICA1 merck-NM_001025598_at ARHGAP30 USF1 merck-NM_001768_at CD8A merck-NM_003978_at PSTPIP1 merck-NM_014716_at ACAP1 merck-AK128740_s_at IL16 merck-NM_006060_a_at IKZF1 merck-BC075820_at IKZF1 merck-NM_016293_at BIN2 merck-NM_012092_at ICOS merck-NM_005442_at EOMES LOC100996624 merck-NM_007074_at CORO1A merck-NM_000206_at IL2RG merck-NM_005041_at PRF1 merck-NM_024898_s_at DENND1C CRB3 merck-NM_173799_at TIGIT merck-NM_001767_at CD2 merck-NM_002348_at LY9 merck-X60502_s_at SPN QPRT merck-NM_153236_at GIMAP7 merck-NM_005601_at NKG7 merck-NM_032496_at ARHGAP9 merck-NM_004877_at GMFG merck-NM_021181_at SLAMF7 merck-NM_018384_at GIMAP5 GIMAP1-GIMAP5 merck-NM_181780_at BTLA merck-NM_001017373_at SAMD3 merck-NM_000734_at CD247 merck-NM_003650_at CST7 merck-NM_172101_at CD8B merck-NM_001803_at CD52 merck-NM_001778_at CD48 merck-NM_001025265_at CXorf65 merck-NM_198929_at PYHIN1 merck-ENST00000379833_at GVINP1 merck-NM_052931_at SLAMF6 merck-NM_001024667_s_at FCRL3 merck-NM_002258_at KLRB1 merck-NM_018556_s_at SIRPG merck-AK090431_s_at NLRC3 merck-NM_018990_at SASH3 XPNPEP2 merck-NM_175900_s_at C16orf54 QPRT merck-ENST00000316577_s_at TESPA1 merck-NM_024070_at PVRIG merck-AY190088_s_at merck-NM_001040067_s_at TRBC2 TRBV3-1 TRBV5-4 TRBV6-5 TRBV7-2 merck-NM_130848_s_at C5orf20 merck-ENST00000381153_at C11orf21 merck-ENST00000382913_s_at TRAC TRAJ17 TRAV20 TRDV2 merck-BC030533_s_at TRBC1 TRBV19 merck-ENST00000244032_a_at ZNF831 merck-ENST00000371030_at ZNF831 merck-ENST00000343625_s_at RASAL3 merck-AF143887_at merck-AK128436_at IKZF3 merck-AI281804_at GPR174 merck-AF086367_at merck-CR598049_at LINC00426 merck-BM700951_at KLRK1 KLRC4-KLRK1 merck-BX648371_at LINC00861 merck-BC070382_at merck2-AW798052_at AKNA merck2-BX640915_at TIGIT merck2-BM678246_at CD37 merck2-NM_025228_at TRAF3IP3 merck2-XM_033379_at WDFY4 merck2-AJ515553_at AMICA1 merck2-BP262340_at IL16 merck2-AK225623_at DENND1C CRB3 merck2-AL833681_at CD96 merck2-BF111803_at ARHGAP15 merck2-BX406128_at CD3G merck2-NM_153701_at merck2-BC020657_at GIMAP4 merck2-AY185344_at PYHIN1 merck2-DR159064_at EOMES LOC100996624 merck2-ENST00000390420_at TRBV3-1 TRBV5-4 TRBV6-5 TRBV7-2 merck2-ENST00000390420_s_at merck2-NM_001010923_at THEMIS merck2-ENST00000390409_at TRBC1 TRBV19 merck2-AX721088_at merck2-ENST00000390393_at TRBV19 merck2-AW341086_at merck2-AA278761_at merck2-AA278761_x_at merck2-ENST00000390394_s_at merck2-AA669142_at merck2-AW007991_at PTPRC merck2-BG743900_at PRKCB merck2-X06318_at PRKCB merck2-BI519527_at IKZF1 merck2-ENST00000390537_s_at merck2-AY292266_x_at merck2-NM_005816_a_at CD96 merck2-NM_198196_a_at CD96 merck2-NM_001114380_x_at ITGAL merck2-NM_007237_a_at SP140 merck2-NM_007237_at SP140 merck2-NM_052931_at SLAMF6 merck2-NM_001558_at IL10RA merck2-NM_007360_at KLRK1 KLRC4-KLRK1 merck2-NM_002209_x_at ITGAL merck2-NM_175900_at C16orf54 QPRT

TABLE 13 Hypoxia signature genes probe Gene merck-NM_002627_at PFKP PITRM1 merck-NM_000302_at PLOD1 merck-NM_001216_at CA9 RMRP merck-ENST00000377093_at KIF1B merck-BC004202_a_at CHEK1 merck-NM_030949_at PPP1R14C merck-CR593119_a_at CLIC4 merck-NM_001255_s_at CDC20 merck-BG679113_s_at KRT6A KRT6B KRT6C merck-NM_002421_at MMP1 merck-BQ217236_a_at SERPINB5 merck-NM_001793_at CDH3 merck-NM_001238_at CCNE1 merck-BU597348_s_at SYNCRIP merck-NM_006516_at SLC2A1 merck-BX648425_a_at DSC2 merck-X15014_a_at RALA merck-NM_018685_at ANLN merck-CR614206_a_at ERO1L merck-NM_001124_at ADM merck-NM_015440_at MTHFD1L merck-ENST00000367307_a_at MTHFD1L merck-NM_058179_at PSAT1 merck-NM_031415_s_at GSDMC merck-NM_005557_x_at KRT16 merck-NM_053016_at PALM2 PALM2-AKAP2 merck-CR602579_a_at CTPS1 merck-NM_001428_s_at ENO1 merck-ENST00000305850_at CENPN CMC2 merck-NM_005978_at S100A2 merck-NM_018643_at TREM1 merck-NM_006505_at PVR merck-NM_080655_s_at MSANTD3 merck-NM_001012507_at CENPW merck-ENST00000258005_a_at NHSL1 merck-AK129763_at LINC00673 merck-XM_927868_s_at PGK1 merck-XM_928117_x_at FAM106B merck-AL359337_at ADM merck-AA148856_s_at SYNCRIP merck2-AI989728_at SERPINB5 merck2-DQ892208_at CA9 RMRP merck2-AK022036_at WWTR1 merck2-AA677426_at merck2-AA677426_s_at merck2-BC004856_at NCS1 merck2-BG252150_at PFKP merck2-BC007633_at AGO2 merck2-BG400371_at merck2-DQ891441_at merck2-NM_017522_AS_at LRP8 merck2-AF039652_at RNASEH1 merck2-AV714642_at ANLN merck2-AB030656_at CORO1C merck2-NM_000291_at PGK1 merck2-NM_005554_at KRT6A merck2-BC002829_at S100A2 merck2-BU681245_at merck2-AK225899_a_at CTPS1 merck2-BC062635_a_at XPO5 merck2-AF257659_a_at CALU merck2-CA308717_at merck2-X56807_at DSC2 merck2-CR936650_at ANLN merck2-AY423725_a_at PGK1 merck2-BC103752_a_at PGK1

TABLE 14 VIM correlated genes probe Gene merck-NM_005211_at CSF1R merck-NM_001699_at AXL merck-NM_032525_at TUBB6 merck-AL710269_a_at CDK14 merck-NM_152653_s_at UBE2E2 merck-NM_032777_s_at GPR124 merck-AF085983_s_at ZEB2 merck-NM_002510_at GPNMB merck-NM_002444_at MSN merck-NM_016938_at EFEMP2 merck-NM_031934_at RAB34 merck-NM_016815_at GYPC merck-NM_005429_at VEGFC merck-NM_003380_a_at VIM merck-ENST00000316623_a_at FBN1 merck-NM_003873_at NRP1 merck-BU625463_s_at EFEMP2 merck-NM_003255_s_at TIMP2 merck-CA447839_at FAM49A merck-AY548106_a_at CCDC80 merck-BC086876_a_at CCDC80 merck-NM_006317_at BASP1 merck-NM_006832_at FERMT2 merck-NM_003118_s_at SPARC merck-NM_005461_at MAFB merck-NM_013352_at DSE merck-NM_002017_at FLI1 merck-NM_020856_at TSHZ3 merck-NM_014737_at RASSF2 merck-NM_014795_at ZEB2 merck-BC025730_at ZEB2 merck-NM_144601_at CMTM3 merck-NM_016429_at COPZ2 merck-NM_012219_s_at MRAS merck-NM_001425_at EMP3 TMEM143 merck-NM_012072_at CD93 merck-NM_016274_s_at PLEKHO1 merck-NM_206853_s_at QKI merck-NM_006868_at RAB31 merck-DB025966_a_at RAB31 merck-AL833176_at CHST11 merck-AF055376_at MAF LOC101928230 merck-CR616358_s_at DCN merck-NM_001031679_at MSRB3 merck-CR604988_a_at CLEC2B merck-NM_015150_at RFTN1 merck-NM_052966_at FAM129A merck-NM_024579_at C1orf54 merck-XM_087386_at HEG1 merck-ENST00000311127_a_at HEG1 merck-ENST00000252031_at C20orf194 merck-ENST00000252032_a_at C20orf194 merck-AK123315_a_at LOC100132891 merck-AK091332_at GNB4 merck2-AF086016_at NRP1 merck2-NM_199511_at CCDC80 merck2-NM_003768_at PEA15 merck2-BC010410_at TIMP2 merck2-BM468535_at merck2-BC023509_at CMTM3 merck2-G43223_a_at VIM merck2-NM_001920_at DCN merck2-NM_015463_at CNRIP1 merck2-CB240675_at merck2-AA664657_x_at VIM merck2-BX352133_s_at merck2-BM754248_at FBN1 merck2-AB266387_s_at CCDC80 merck2-AK075210_a_at CCDC80 merck2-CX871427_at BASP1 merck2-DQ892556_a_at DCN LOC101928584 merck2-BQ632060_x_at VIM merck2-BM999558_x_at VIM

TABLE 15 CDH1 correlated genes probe Gene merck-NM_002773_at PRSS8 merck-NM_020770_at CGN merck-M34309_a_at ERBB3 merck-NM_002273_x_at KRT8 merck-NM_004360_at CDH1 TANGO6 merck-NM_024729_s_at MYH14 KCNC3 merck-NM_052886_at MAL2 merck-BC069241_a_at ESRP2 merck-NM_002670_at PLS1 merck-NM_004433_a_at ELF3 merck-ENST00000367284_at ELF3 merck-NM_001034915_s_at ESRP1 merck-BC016153_s_at TMEM45B merck-BX364926_at IRF6 merck-NM_006147_at IRF6 merck-ENST00000378957_a_at EPCAM merck-NM_001305_at CLDN4 merck-NM_007183_at PKP3 merck-NM_001008844_at DSP merck-NM_020387_at RAB25 merck-NM_173853_s_at KRTCAP3 merck-NM_005498_at AP1M2 merck-NM_199187_x_at KRT18 merck-NM_001017967_at MARVELD3 PHLPP2 merck-NM_000346_at SOX9 merck-NM_024320_at PRR15L merck-NM_001307_at CLDN7 merck-NM_144724_at MARVELD2 merck-NM_173481_t MISP merck-AK093149_a_at MYO5B merck-AK026517_at EHF merck-CB160685_s_at HNF4A merck-AF086028_at ERBB3 merck2-NM_001982_at ERBB3 merck2-AI052130_at TMEM45B merck2-CK818800_at ESRP1 merck2-AB209992_at DSP merck2-CN341876_at IRF6 GRM7 merck2-NM_002354_at EPCAM merck2-NM_001305_at CLDN4 merck2-NM_199187_x_at merck2-NM_001307_at CLDN7 merck2-BE542388_at CDH1 TANGO6 merck2-AK025901_a_at ESRP2 merck2-CA314539_at NFATC3 merck2-BM981128_at merck2-ENST00000367021_at IRF6 merck2-AJ011497_a_at CLDN7 merck2-NM_182517_at C1orf210

TABLE 16 Prognosis component 1 (prg1) genes Probe Gene merck-NM_001192_at TNFRSF17 merck-NM_144646_at IGJ merck2-AF343666_at merck2-DQ884395_a_at IGJ merck-NM_016459_at MZB1 merck2-AK125079_s_at merck2-BX648616_s_at merck-NM_006235_at POU2AF1 merck-AX747748_s_at IGHA1 IGHA2 IGH merck2-BC020889_at IGJ merck2-BF174271_at MZB1 merck-NM_001783_at CD79A merck2-BC007782_at IGLC1 merck2-U52682_at IRF4 merck-NM_006875_at PIM2 merck-ENST00000290730_s_at DERL3 merck2-ENST00000304187_x_at merck2-ENST00000390629_x_at merck-ENST00000379877_x_at IGHA1 IGHG1 IGH merck2-ENST00000390243_x_at merck-AF343662_at FCRL5 merck2-ENST00000390290_x_at merck-BC070352_x_at IGLV3-21 merck2-XM_037686_at DERL3 merck-ENST00000241813_at TNFRSF17 merck-NM_014879_at P2RY14 merck2-ENST00000390273_x_at IGKC IGKV1-16 IGKV1D-16 merck2-ENST00000390243_at merck-NM_017709_at FAM46C merck2-DB327580_at FCRL5 merck2-ENST00000379900_x_at merck2-ENST00000390290_at merck-AF035036_x_at IGK IGKV3-20 IGKV3D-20 merck-BC042060_x_at LOC100509541 merck2-ENST00000390615_x_at merck2-L37307_x_at merck-ENST00000333289_x_at IGLV6-57 merck-U07440_x_at OR6C4 IGKV3-11 IGKV3D-11 IGKV3D-20 RHNO1 merck-AK091834_at FENDRR merck-X57809_x_at merck2-ENST00000390615_at merck2-U07440_x_at merck2-ENST00000390630_x_at merck-AK024399_at TSPAN11 merck2-CD703280_at IGKC IGK IGKV3-11 IGKV3-20 IGKV3D-20 merck2-BE935035_at merck2-NM_017773_at LAX1 merck-NM_001242_at CD27 merck-ENST00000360329_at KIAA0125 merck2-ENST00000359488_x_at IGKC IGKV1D-39 IGKV1-39 merck2-ENST00000390272_x_at IGKV1D-17 merck2-Z47250 x_at merck-NM_017773_at LAX1 merck-CR605298_s_at FENDRR merck2-AF408729_x_at IGKC IGKV2-30 IGKV2D-30 merck-NM_002460_at IRF4 merck-ENST00000382880_x_at CYAT1 IGLL5 IGLC1 IGLC2 IGLC3 IGLJ3 IGLV1- 44 IGLV3-25 IGLV4-3 merck2-S67637_x_at merck2-AF035036_x_at IGKV3-20 merck-ENST00000304187_x_at IGK IGKV1-5 IGKV3-15 IGKV3D-15 merck2-ENST00000390299_x_at IGLV1-40 IGLV5-39 merck-BC022823_x_at IGLV3-25 merck-NM_014792_at KIAA0125 merck2-BC022823_x_at IGLV3-25 merck-NM_003037_at SLAMF1 merck-NM_021181_at SLAMF7 merck-NM_031281_at FCRL5 merck-NM_001775_at CD38 merck-NM_000036_at AMPD1 merck2-ENST00000390276_x_at merck2-ENST00000390285_at IGLV6-57 merck-ENST00000358611_x_at IGKC IGKV1D-16 merck-DB350188_a_at IGHG1 IGHG3 IGHM merck-NM_001002862_at DERL3 SMARCB1 merck-AI676062_at TCONS_00024492 LOC101928582 LOC146513 TCONS_00024764 merck-AJ004955_at IGKV4-1 merck2-BC009851_at IGHM merck-AK097071_s_at IGHM merck-AAS02609_a_at TRPA1 merck2-CR749861_x_at merck2-ENST00000390265_x_at IGKC IGKV1-33 IGKV1D-33 merck-NM_145285_s_at NKX2-3 merck-NM_020939_at CPNE5 merck2-M34461_at CD38 merck2-ENST00000379894_x_at merck-ENST00000331195_x_at merck-NM_002986_s_at CCL11 merck2-S67987_x_at merck2-AF076199_at merck2-XM_001133802_at LOC101928582 TCONS_00024492 LOC146513 TCONS_00024764 merck-ENST00000359488_x_at IGKV1D-39 IGKV@ IGKV1-39 merck-X57817_x_at IGLJ3 merck2-AF076199_x_at merck-ENST00000379884_x_at IGHG1 IGHV1-46 merck-L43092_x_at CKAP2 IGLJ3 IGLV3-19 merck-BX648045_s_at ANKRD36B merck2-BC017850_at CCL11 merck-NM_030764_s_at FCRL2 merck2-ENST00000390593_at IGHM IGHV6-1 merck2-Z14216_x_at IGHV3-15

TABLE 17 Prognosis component 2 (prg2) genes probe Gene merck-NM_001017962_at P4HA1 merck2-BX648829_at P4HA1 merck2-DQ892544_at SPP1 merck2-AK124671_a_at TMCC1 merck-BC039859_a_at TMCC1 merck2-BM985119_a_at VEGFA merck-NM_000582_at SPP1 merck-ENST00000373907_a_at DLGAP4 merck-ENST00000199940_a_at MAP2 merck-AK021681_a_at SEPT10 merck2-Z29328_a_at UBE2H merck-BP311362_a_at LUZP6 MTPN merck-NM_181552_at CUX1 merck-AF125392_a_at INSIG2 merck2-BE900907_a_at UBE2H merck-NM_054034_a_at FN1 merck-NM_199235_at COLEC11 merck-X54315_a_at CDH2 merck2-BQ277651_at CDH2 merck-AK125666_a_at VEGFA merck-NM_002182_at IL1RAP merck2-AF277174_at EGLN1 merck-AF028828_at SNTB1 merck-DA993973_a_at KBTBD2 merck-ENST00000377499_a_at LMO7 merck-BF056045_a_at MPRIP merck-CR612713_s_at MAPK14 merck-AK056350_s_at DCBLD2 merck2-AI765059_at MPRIP merck2-CB115148_at PLIN2 merck-ENST00000367307_a_at MTHFD1L merck2-NM_133376_a_at ITGB1 merck-BG706780_s_at RHEB merck2-BG699831_at INSIG2 merck-ENST00000369578_a_at ZNF292 merck2-DB483456_at YWHAG merck-NM_053043_at RBM33 merck-NM_022347_at TOR1AIP2 merck2-BX647140_at DCBLD2 merck2-AA446940_at DLGAP4 merck-BU538528_s_at MAP2 merck2-DB498046_x_at HSP90AB1 merck-BC010860_a_at SERPINE1 merck-ENST00000382881_a_at ZMYM2 merck2-S42303_at CDH2 merck-AK125700_a_at PLOD2 merck2-BQ000301_at NAB1 LOC101927315 merck-NM_177444_s_at PPFIBP1 merck-M94010_a_at F5 merck-AK057337_at LINC00924 merck2-BE669868_a_at ANKLE2 merck-ENST00000376200_s_at NALCN merck2-AF322916_at UACA LOC101929151 merck-BQ440605_a_at ITGB1 merck-DB226799_a_at PTK2 merck-NM_006516_at SLC2A1 merck-CR624299_s_at GRB10 merck-AK000990_a_at UACA merck2-NM_178826_at ANO4 UTP20 merck-NM_005401_at PTPN14 merck-BX640712_a_at TMCC1 merck-BX451561_a_at ARHGEF7 merck-AF075090_a_at MET merck-BI917224_a_at PLIN2 merck-DA409370_a_at MAP4K3 merck2-AW162846_at merck-NM_001084_at PLOD3 merck2-CA423142_a_at MLLT4 KIF25 merck2-DB498046_at HSP90AB1 merck2-NM_000908_at NPR3 merck-NM_015852_at ZNF117 merck-NM_000908_at NPR3 merck-NM_001792_a_at CDH2 merck2-BC018124_at HSPH1 merck-NM_021175_at HAMP merck-BC065279_a_at IWS1 merck-BC001136_a_at PLEKHA1 merck-AV717806_a_at HSPH1 merck2-M16967_at F5 merck-NM_018433_s_at KDM3A merck2-BQ217998_a_at ANKLE2

TABLE 18 Prognosis component 3 genes probe Gene merck-NM_001013029_at IGFBP1 merck-BG567539_a_at FGA merck2-NM_021871_at FGA merck2-BC106760_at FGB merck-NM_005141_at FGB merck2-AI174982_at FGB merck-NM_000509_at FGG merck2-NM_021870_at FGG merck-NM_002216_at ITIH2 merck2-BC007058_at APCS merck-NM_001639_at APCS merck2-NM_000567_at CRP merck-NM_000567_at CRP merck-NM_000583_at GC merck2-AV645562_a_at ALB merck2-U22961_a_at ALB merck2-AF119840_at ALB merck2-DQ891414_x_at ALB merck2-AY960291_x_at ALB

Example 4 Prognostic Model for Kidney Cancer

This example describes a kidney cancer prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

A total of 893 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model was validated using the second half of samples. In the first half of samples, 443 samples had outcome data (live or death). In the second half of samples, 444 had outcome data. The detailed last follow-up dates for the good outcome patients are incomplete. In the first half of samples, 106 out of 283 good outcome patients did not have the last follow-up date. In the second half of samples, 146/315 good outcome patients did not have the last follow-up date. In poor outcome patients, all but one had last follow-up dates.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 443 training samples which are either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 22 & 23. Genes in Table 23 are highly enriched for cell cycle and cell proliferation pathways.

TABLE 22 Prognosis signature component 1 (anti-correlated with poor outcome) genes probe Gene merck-NM_000901_at NR3C2 merck-M13994_a_at BCL2 merck2-BM977883_at FAM221B merck-NM_021117_at CRY2 merck-NM_001280_a_at CIRBP merck2-BC036093_at HLF merck-NM_018945_s_at PDE7B merck-NM_138333_at FAM122A merck-BQ709647_a_at HLF merck-NM_014014_at SNRNP200 merck2-AF316873_at PINK1 DDOST merck-H05603_a_at THRA NR1D1 merck2-NM_182517_at C1orf210 merck2-AB075482_at merck2-BF433548_at merck2-NM_003250_at merck-NM_025202_at EFHD1 merck-NM_182517_at C1orf210 merck2-CK005338_at merck-ENST00000375138_s_at MINOS1 merck2-NM_003250_a_at THRA NR1D1 merck-ENST00000377991_at TMEM8B FAM221B merck-ENST00000269197_at ASXL3 merck2-BG674122_a_at HLF merck-ENST00000264431_s_at RAPGEF2 merck-NM_014234_a_at HSD17B8 merck-NM_015316_at PPP1R13B merck2-BU159596_at BCL2 merck-NM_024563_at NPR3 merck-ENST00000307249_at EPB41L4A-AS2 merck-NM_000633_at BCL2 merck-AY117034_a_at EMX2OS merck-NM_201536_s_at NDRG2 merck-NM_175709_at CBX7 merck2-BF940198_at LIFR-AS1 LIFR merck-AJ315514_a_at NR3C2 merck-NM_002126_at HLF merck2-AF070541_at LOC284244 merck-BX335786_s_at FAM47E merck-AK126966_at TADA2B merck2-BC128418_at CBX7 merck-BC063296_at MTMR10 FAN1 merck2-BX408834_at NDRG2 merck-NM_080597_at OSBPL1A merck2-AK021580_at PPP1R13B merck-NM_014828_at TOX4 METTL3 merck-NM_017719_at SNRK merck-NM_032385_at FAXDC2 merck2-AW612403_at CCDC176 ALDH6A1 merck-BX437500_at SCAI merck-NM_000908_at NPR3 merck-NM_145689_s_at APBB1 SMPD1 merck-NM_004928_at C21orf2 merck2-NM_030807_at SLC2A11 merck2-AI927896_at merck-BG536817_a_at TMEM245 merck2-NM_000908_at NPR3 merck-NM_001042_at SLC2A4 merck-ENST00000332811_at ZNRF3 merck-NM_024900_at PHF17 merck-AK091971_a_at PKHD1 merck-NM_006393_at NEBL merck-NM_031889_at ENAM merck-AK021616_at OTUD7A merck-BC038509_a_at RCAN2 merck-AK123831_at CDS2 merck2-NM_003991_at EDNRB merck-ENST00000344980_s_at ZNF433 merck2-DQ890997_a_at APBB1 merck-NM_013381_at TRHDE merck-AK001936_a_at EIF4EBP2 merck-BC095414_a_at BDH2 merck-NM_032717_at AGPAT9 merck-ENST00000377448_a_at ZNF204P merck-AK021522_a_at VAMP2 merck2-AW966622_at NEBL merck2-ENST00000377187_at NEBL merck-BC014248_a_at TMEM245 merck-AB007969_at CLMN merck-NM_001979_at EPHX2 merck-BM925725_a_at LIFR merck-NM_153281_s_at HYAL1 merck2-AA043801_at SYNJ2BP merck-NM_032233_at SETD3 BCL11B merck-NM_004098_s_at EMX2 merck2-BF945736_at C21orf2 merck2-XM_085862_s_at ILF3-AS1 merck-DA383742_a_at EMX2OS merck-NM_182758_at WDR72 merck2-NM_023926_a_at ZSCAN18 merck-BC042390_s_at VTI1B merck-NM_021229_at NTN4 merck-NM_152444_at PTGR2 merck2-BU687744_at merck-NM_020698_at TMCC3 merck2-BC032376_at PHF17 merck-NM_030911_at CDADC1 merck2-AI761584_at merck2-BC034387_at SLC2A4 merck-AK055143_s_at

TABLE 23 Prognosis signature component 2 (correlated with poor outcome) genes probe Gene merck2-AF043294_at BUB1 RGPD6 merck-NM_004336_at BUB1 RGPD6 merck-NM_005733_at KIF20A CDC23 merck2-NM_005196_at CENPF merck-NM_012112_at TPX2 merck-NM_181802_at UBE2C merck-NM_001809_at CENPA merck2-BC006325_at GTSE1 TRMU merck-NM_004701_at CCNB2 merck2-AF098158_at TPX2 merck2-BC006325_x_at GTSE1 TRMU merck-NM_001786_a_at CDK1 RHOBTB1 merck-ENST00000243201_a_at HJURP merck-NM_001255_s_at CDC20 merck-NM_004219_x_at PTTG1 merck2-BC034607_at ASPM merck2-BC098582_at KIF14 merck2-AV714642_at ANLN merck-NM_018131_at CEP55 merck-NM_002497_at NEK2 merck-NM_001067_at TOP2A merck-NM_018685_at ANLN merck-BC075828_a_at GTSE1 merck-NM_031299_at CDCA3 GNB3 merck2-BC107750_at CDK1 RHOBTB1 merck-NM_004217_at AURKB merck2-NM_018410_at HJURP merck-CR596700_a_at RRM2 merck-NM_016343_at CENPF merck-BI868409_a_at MKI67 merck2-CR936650_at ANLN merck-BF511624_s_at BUB1B merck-NM_018101_at CDCA8 merck-U63743_a_at KIF2C merck2-NM_145060_a_at SKA1 merck2-BC001651_at CDCA8 merck-NM_001211_at BUB1B merck-NM_012484_at HMMR merck-NM_014750_at DLGAP5 merck-NM_018136_s_at ASPM merck2-NM_031966_at CCNB1 merck-NM_021953_at FOXM1 merck2-AL519719_a_at BIRC5 merck-NM_130398_at EXO1 merck-NM_014176_at UBE2T merck-NM_005030_at PLK1 merck-NM_145060_at SKA1 merck2-AL517462_s_at merck-NM_145697_at NUF2 merck-NM_016426_at GTSE1 TRMU merck-NM_153824_a_at PYCR1 merck2-NM_001168_at BIRC5 merck2-NM_001039535_a_at SKA1 merck-NM_017947_at MOCOS merck-NM_152515_at CKAP2L merck-ENST00000333706_x_at BIRC5 merck-NM_003318_at TTK merck-AK223428_a_at BIRC5 merck-AK024080_a_at TOP2A merck-NM_002466_at MYBL2 merck-NM_005480_at TROAP merck2-ENST00000370966_a_at DEPDC1 OTUD7A merck-NM_080668_at CDCA5 merck-ENST00000335534_s_at KIF18B merck2-ENST00000372927_at CENPI merck2-BX349325_at PRR11 merck-BF308644_s_at CENPI merck-NM_012310_at KIF4A GDPD2 merck-NM_018304_s_at PRR11 merck-NM_001790_at CDC25C merck-CR602926_s_at CCNB1 merck2-ENST00000333706_s_at merck-NM_002417_at MKI67 merck2-NM_145061_at SKA3 merck-NM_182513_at SPC24 merck-NM_019013_at FAM64A PITPNM3 merck2-NM_001761_at CCNF merck2-BT006759_at KIF2C merck-NM_004237_at TRIP13 merck-NM_152463_s_at EME1 merck-NM_014791_at MELK merck-NM_005192_at CDKN3 merck-AK055931_a_at SHCBP1 merck-NM_018234_at STEAP3 merck-AF331796_a_at NCAPG merck-NM_152259_s_at TICRR KIF7 merck-NM_198436_s_at AURKA merck2-AL832036_at CKAP2L merck2-AK097710_at CDC25C merck2-NM_017779_at DEPDC1 merck2-NM_024745_at SHCBP1 merck-NM_001813_at CENPE merck2-BG497357_at NUF2 merck-NM_199413_at PRC1 merck-hCT1776373.2_s_at DEPDC1 OTUD7A merck-BC048988_a_at SKA3 merck2-DQ892840_a_at CDC6 merck-NM_018248_at NEIL3 merck-NM_001237_a_at CCNA2 EXOSC9 merck-NM_033300_at LRP8

A kidney cancer risk model was built from the training set using a general linear model (from the R package) using the following equation:


Kidney Cancer Risk Score=1.54563−(0.19522*prg1)+(0.06519*prg2)   (Formula 7),

where “prg1” is a score calculated from the prognosis genes in Table 22 and “prg2” is a score calculated from prognosis genes in Table 23. These scores are calculated by averaging the log2(intensity) of each probe in the geneset.

The performance of this model was evaluated in reserved validation set of 444 samples. FIG. 14 shows the predicted death rate vs. the actual average (running average of 100 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 24.

TABLE 24 Average death rate versus prediction score. Prediction score Number of samples Number of deaths Rate <0.2 138 22 0.15942029 0.2-0.3 109 22 0.201834862 0.3-0.4 56 13 0.232142857 0.4-0.5 33 10 0.303030303 0.5-0.6 33 16 0.484848485 0.6-0.7 29 13 0.448275862 >0.7 46 33 0.717391304

Using a threshold of 0.4, the odds ratio for overall survival was 4.5 (95% CI: 2.9-7.0), Fisher's Exact Test p-value=1.2×10−11.

Patients can be further divided into good (risk score<0.35), medium (score 0.35-0.6) and poor (score>0.6) prognosis groups. FIG. 15 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 62.7 (P=2.4×10−14).

The number of genes in each pathway was reduced to 10 genes.

Prognosis Signature Component 1 (prg1):

    • Probe IDs: merck-NM_021117_at, merck-NM_000901_at, merck2-BC036093_at, merck-AY117034_a_at, merck2-BM977883_at, merck2-NM_020139_at, merck-M13994 a_at, merck2-NM_001608_at, merck-NM_201536_s_at, merck-NM_024563_at
    • Gene symbols: CRY2, NR3C2, HLF, EMX2OS, FAM221B, BDH2, BCL2, ACADL, NDRG2, NPR3

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck-NM_012112_at, merck-NM_004701_at, merck-NM_004217_at, merck-ENST00000243201_a_at, merck-NM_001809_at, merck2-NM_005196_at, merck-NM_145060_at, merck-NM_018131_at, merck-NM_004219 x at, merck-NM_021953_at
    • Gene symbols: TPX2, CCNB2, AURKB, HJURP, CENPA, CENPF, SKA1, CEP55, PTTG1, FOXM1

The scores derived from these 10-genes correlated to the original scores at the level of 0.97 for prg1 and 0.99 for prg2.

Using the reduced gene sets, the updated predictive model is:


Kidney Cancer Risk Score=0.65473+(−0.10355*prg1)+(0.08053*prg2)   (Formula 8).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

FIG. 16 shows the predicted death rate vs. the actual average (running average of 100 samples as ranked by the prediction score) death rate for this updated model. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 25.

TABLE 25 Average death rate versus prediction score. Prediction score Number of samples Number of deaths Rate <0.2 126 20 0.158730159 0.2-0.3 121 26 0.214876033 0.3-0.4 58 15 0.25862069 0.4-0.5 39 11 0.282051282 0.5-0.6 28 11 0.392857143 0.6-0.7 26 15 0.576923077 >0.7 46 31 0.673913043

Using a threshold of 0.42, the odds ratio for overall survival was 4.4 (95% CI: 2.8-6.9), Fisher's Exact Test p-value=4.3×10−11.

Patients can be further divided into good (risk score<0.35), medium (score 0.35-0.6) and poor (score>0.6) prognosis groups. FIG. 17 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 68.4 (P=1.4×10−15).

Example 5 Prognostic Model for Brain Cancer

This example describes a brain cancer prognosis model based on gene expression profiling data. The model contains three gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

A total of 517 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model validated using the second half of samples. In the first half of samples, 257 samples had outcome data (live or death). In the second half of samples, also 257 had outcome data. The detailed last follow-up dates for the good outcome patients was incomplete. In the first half of samples, 32 out of 95 good outcome patients did not have the last follow-up date. In the second half of samples, 49/121 good outcome patients did not have the last follow-up date. In poor outcome patients, training and validation set each had one without the last follow-up date.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 257 training samples which were either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 26 & 27. Genes in Table 27 are highly enriched for cell cycle and cell proliferation pathways.

TABLE 26 Prognosis signature component 1 (anti-correlated with poor outcome) genes probe Gene merck-NM_021117_at CRY2 merck-NM_152754_at SEMA3D merck2-NM_001329_at CTBP2 merck-NM_014912_at CPEB3 merck-NM_004962_at GDF10 merck2-BF055210_a_at CTBP2 merck-ENST00000369884_at CYP17A1-AS1 merck-NM_002126_at HLF merck2-BM975249_at SGMS1 merck-ENST00000344293_s_at TAF3 merck-AK026683_a_at SGMS1 merck2-NM_001047160_at NET1 merck-BM450726_at ZRANB1 merck2-NM_004657_at SDPR merck-ENST00000308281_a_at NET1 merck-NM_001010888_s_at ZC3H12B merck2-AW591673_at merck-BQ709647_a_at HLF merck-NM_147156_at SGMS1 merck2-BC036093_at HLF merck-BC035870_a_at MIPOL1 merck2-AK125919_at SCAPER merck2-DB321909_at SYT15 merck2-BM728590_at SESN1 merck-NM_173576_s_at MKX merck-BC016475_a_at SDPR merck2-BF055210_at merck2-BG674122_a_at HLF merck2-BM555890_a_at SDPR merck-BC036444_a_at CPEB3 merck-ENST00000374390_s_at 8-Mar merck-NM_144591_a_at C10orf32 merck2-BM728590_a_at SESN1 merck-ENST00000335753_at merck-AK123201_at MTMR7 VPS37A merck-NM_001609_at ACADSB merck2-R56002_at TTC33 merck-NM_019036_s_at HMGCLL1 merck2-ENST00000379483_at merck2-ENST00000308161_at HMGCLL1 merck-ENST00000368886_at IKZF5 merck-AK026718_at SNX2 merck-NM_203441_at FRA10AC1 merck-NM_138731_at MIPOL1 merck-NM_031469_at SH3BGRL2 merck2-AL832477_at C10orf32 merck-NM_022117_at TSPYL2 merck-NM_003939_at BTRC merck2-AL834189_at VPS37A MTMR7 merck-CR598481_at TTC33 merck2-DQ269985_at AKR1C3 merck-AV654599_s_at AKR1C3 merck2-NM_031912_at merck2-CR593590_at GNAL MPPE1 merck-NM_000997_at RPL37 merck2-AL136713_a_at GHITM merck-NM_014454_s_at SESN1 merck-NM_021785_at RAI2 merck-NM_017580_a_at ZRANB1 merck-AK001299_at VWF merck-ENST00000346874_at PARD3 merck2-AB188491_at OTUD1 merck2-Y07511_at OAT merck-NM_006624_at ZMYND11 merck-NM_153277_at SLC22A6 CHRM1 merck2-DA751278_at RPL13 merck-AK122845_a_at GABRG1 merck2-BC050310_at CCNY merck-ENST00000330762_at NUTM2D merck-AY491432_at merck-AK022354_at METTL10 merck2-NM_130439_at MXI1 merck-NM_012141_at INTS6 merck-ENST00000355854_at CAB39L merck-ENST00000369203_at SLC18A2 merck-NM_003216_at TEF merck-BX366291_at merck2-W94048_at TIAL1 merck-NM_024701_at ASB13 merck-NM_152503_at MROH8 merck-ENST00000268533_at NUDT7 merck2-C04536_a_at MXI1 merck-DA165254_a_at CACNA2D3 merck-NM_175607_at CNTN4 merck-AW959468_s_at merck2-AI003348_at NMNAT2 merck-NM_022039_at FBXW4 merck2-XM_001127131_at NUDT7 merck-ENST00000369895_a_at ARL3 merck2-AI192627_at PPP3CB merck2-BC035128_a_at MXI1 merck-NM_032138_at KBTBD7 merck-ENST00000369619_a_at MXI1 merck-NM_016929_at CLIC5 merck-ENST00000298035_at OTUD1 merck-NM_021132_at PPP3CB merck-CB048235_at merck2-AA815447_at CACNA2D3 merck2-BF248252_at merck-NM_001050_at SSTR2

TABLE 27 Prognosis signature component 2 (correlated with poor outcome) genes probe Gene merck-CR596700_a_at RRM2 merck2-AL517462_s_at merck-NM_145060_at SKA1 merck-NM_198436_s_at AURKA merck2-NM_001039535_a_at SKA1 merck2-NM_145060_a_at SKA1 merck-ENST00000333706_x_at BIRC5 merck-AK223428_a_at BIRC5 merck-NM_004219_x_at PTTG1 merck-NM_012310_at KIF4A GDPD2 merck-NM_001809_at CENPA merck2-ENST00000333706_s_at merck-NM_001276_at CHI3L1 merck-NM_018101_at CDCA8 merck-ENST00000360566_at RRM2 merck2-BC001651_at CDCA8 merck2-AF098158_at TPX2 merck-NM_012112_at TPX2 merck-NM_005733_at KIF20A CDC23 merck-U63743_a_at KIF2C merck2-AK123247_at MYH11 NDE1 merck2-ENST00000331944_s_at merck-NM_181802_at UBE2C merck2-NM_018410_at HJURP merck2-BT006759_at KIF2C merck2-M87338_at RFC2 merck-NM_152637_at METTL7B ITGA7 merck-NM_182513_at SPC24 merck-NM_018154_at ASF1B PRKACA merck2-AL519719_a_at BIRC5 merck2-BC007417_at POC1A merck-NM_021953_at FOXM1 merck-NM_016426_at GTSE1 TRMU merck-CR602926_s_at CCNB1 merck-NM_014791_at MELK merck-NM_006342_at TACC3 merck-NM_004701_at CCNB2 merck-NM_004217_at AURKB merck-NM_144569_s_at SPOCD1 merck2-NM_001168_at BIRC5 merck2-BC006325_at GTSE1 TRMU merck-NM_018131_at CEP55 merck-AY605064_at CLSPN merck-NM_004336_at BUB1 RGPD6 merck-NM_031299_at CDCA3 GNB3 merck2-AF043294_at BUB1 RGPD6 merck2-NM_014397_at NEK6 merck-NM_001255_s_at CDC20 merck2-ENST00000370966_a_at DEPDC1 OTUD7A merck-ENST00000243201_a_at HJURP merck-NM_003258_at TK1 merck-CR602847_a_at KIAA0101 merck-NM_006547_at IGF2BP3 AMOTL1 MALSU1 merck2-BC006325_x_at GTSE1 TRMU merck-BC075828_a_at GTSE1 merck-NM_014750_at DLGAP5 merck-NM_203394_at E2F7 merck-ENST00000308604_s_at LINC00152 MIR4435-1HG merck-AF469667_a_at MLF1IP merck-BI868409_a_at MKI67 merck-NM_016639_at TNFRSF12A CLDN9 merck-CR607300_a_at MKI67 merck-NM_001237_a_at CCNA2 EXOSC9 merck-NM_152515_at CKAP2L merck-AK055931_a_at SHCBP1 merck-NM_005192_at CDKN3 merck2-AK000490_a_at DEPDC1 merck-NM_012291_at ESPL1 PFDN5 merck-BC106033_s_at SMC4 merck2-BC034607_at ASPM merck-NM_152562_s_at CDCA2 merck-NM_004237_at TRIP13 merck2-AK026140_at merck-NM_001813_at CENPE merck2-BC005978_at KPNA2 merck2-NM_024745_at SHCBP1 merck-CR610123_a_at POC1A merck-NM_001790_at CDC25C merck2-Y00472_a_at SOD2 merck2-BC025232_at CDC6 merck2-NM_017779_at DEPDC1 merck-NM_004526_at MCM2 merck2-BC107750_at CDK1 RHOBTB1 merck-BX649059_at GAS2L3 merck-NM_005480_at TROAP merck-NM_007243_a_at NRM merck2-NM_031966_at CCNB1 merck-NM_001024466_s_at SOD2 merck2-BC005978_s_at KPNA2 merck-NM_080668_at CDCA5 merck-NM_004911_at PDIA4 merck-BC004202_a_at CHEK1 merck-NM_003504_at CDC45 merck2-BC098582_at KIF14 merck2-M36693_a_at SOD2 merck-NM_012145_a_at DTYMK merck-NM_017581_at CHRNA9 merck2-BM464374_at CENPE merck-NM_001845_at COL4A1 merck2-DQ890621_at CDC45

TABLE 28 Hypoxia signature probe Gene merck-NM_002627_at PFKP PITRM1 merck-NM_000302_at PLOD1 merck-NM_001216_at CA9 RMRP merck-ENST00000377093_at KIF1B merck-BC004202_a_at CHEK1 merck-NM_030949_at PPP1R14C merck-CR593119_a_at CLIC4 merck-NM_001255_s_at CDC20 merck-BG679113_s_at KRT6A KRT6B KRT6C merck-NM_002421_at MMP1 merck-BQ217236_a_at SERPINB5 merck-NM_001793_at CDH3 merck-NM_001238_at CCNE1 merck-BU597348_s_at SYNCRIP merck-NM_006516_at SLC2A1 merck-BX648425_a_at DSC2 merck-X15014_a_at RALA merck-NM_018685_at ANLN merck-CR614206_a_at ERO1L merck-NM_001124_at ADM merck-NM_015440_at MTHFD1L merck-ENST00000367307_a_at MTHFD1L merck-NM_058179_at PSAT1 merck-NM_031415_s_at GSDMC merck-NM_005557_x_at KRT16 merck-NM_053016_at PALM2 PALM2-AKAP2 merck-CR602579_a_at CTPS1 merck-NM_001428_s_at ENO1 merck-ENST00000305850_at CENPN CMC2 merck-NM_005978_at S100A2 merck-NM_018643_at TREM1 merck-NM_006505_at PVR merck-NM_080655_s_at MSANTD3 merck-NM_001012507_at CENPW merck-ENST00000258005_a_at NHSL1 merck-AK129763_at LINC00673 merck-XM_927868_s_at PGK1 merck-XM_928117_x_at FAM106B merck-AL359337_at ADM merck-AA148856_s_at SYNCRIP merck2-AI989728_at SERPINB5 merck2-DQ892208_at CA9 RMRP merck2-AK022036_at WWTR1 merck2-AA677426_at merck2-AA677426_s_at merck2-BC004856_at NCS1 merck2-BG252150_at PFKP merck2-BC007633_at AGO2 merck2-BG400371_at merck2-DQ891441_at merck2-NM_017522_AS_at LRP8 merck2-AF039652_at RNASEH1 merck2-AV714642_at ANLN merck2-AB030656_at CORO1C merck2-NM_000291_at PGK1 merck2-NM_005554_at KRT6A merck2-BC002829_at S100A2 merck2-BU681245_at merck2-AK225899_a_at CTPS1 merck2-BC062635_a_at XPO5 merck2-AF257659_a_at CALU merck2-CA308717_at merck2-X56807_at DSC2 merck2-CR936650_at ANLN merck2-AY423725_a_at PGK1 merck2-BC103752_a_at PGK1

The prognosis model was built in the training set using a general linear model (from the R package) using the following equation:


Brain Cancer Risk Score=−0.28894+(−0.12713*prg1)+(0.09353*prg2)+(0.15399*hscore)   (Formula 9),

where “prg1” is a score calculated from prognosis genes in Table 26, “prg2” is a score calculated from prognosis genes in Table 27, and “hscore” is a hypoxia pathway score calculated from genes in Table 28. The scores can be calculated by averaging the log2(intensity) of each probe in the geneset.

The performance of this model was evaluated in reserved validation set of 257 samples. FIG. 18 shows the predicted death rate vs. the actual average (running average of 100 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 29.

TABLE 29 Average death rate versus prediction score. Prediction score Number of samples Number of deaths Rate <0.3 57 9 0.157894737 0.3-0.5 35 14 0.4 0.5-0.7 30 17 0.566666667 0.7-0.9 83 58 0.698795181 >0.9 52 38 0.730769231

Using a threshold of 0.58, the odds ratio for overall survival was 6.3 (95% CI: 3.6-10.9), Fisher's Exact Test p-value=1.5×10−11.

Patients can be further divided into good (risk score<0.4), medium (score 0.4-0.75) and poor (score>0.75) prognosis groups. FIG. 19 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 57.5 (P=3.2×10−13).

The number of genes in each pathway was reduced to 10 genes.

Prognosis Signature Component 1 (prg1):

    • Probe IDs: merck-NM_002126_at, merck2-BF055210_a_at, merck-NM_014912_at, merck2-BM975249_at, merck2-NM_001329_at, merck-BM450726_at, merck-NM_003939_at, merck-NM_001609_at, merck-NM_001010888_s_at, merck-ENST00000380064 at
    • Gene symbols: HLF, CTBP2, CPEB3, SGMS1, CTBP2, ZRANB1, BTRC, ACADSB, ZC3H12B, REPS2

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck-NM_145060_at, merck-NM_012112_at, merck-NM_004701_at, merck-NM_001809_at, merck-ENST00000333706_x_at, merck-CR596700_a_at, merck-NM_198436_s_at, merck-NM_004217_at, merck-U63743_a_at, merck2-BC001651_at
    • Gene symbols: SKA1, TPX2, CCNB2, CENPA, BIRC5, RRM2, AURKA, AURKB, KIF2C, CDCA8

Hypoxia Signature:

    • Probe IDs: merck-NM_018643_at, merck-BC010860_a_at, merck-NM_013332_at, merck-X15014_a_at, merck-NM_001625_a_at, merck-NM_001024466_s_at, merck2-BQ015108_at, merck2-BC103752_a_at, merck-NM_001039667_s_at, merck2-NM_001042422_at
    • Gene symbols: TREM1, SERPINE1, HILPDA, RALA, AK2, SOD2, ARL4C, PGK1, ANGPTL4, SLC16A3

The scores derived from these 10-genes are correlated to the original scores at the level of 0.97 for prgl, 0.98 for prg2 and 0.84 for the hypoxia signature.

Using the reduced gene sets, the updated predictive model is:


Brain Cancer Risk Score=−1.320607+(−0.003094*prg1)+(0.094341*prg2)+(0.143865*hscore)   (Formula 10).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

FIG. 20 shows the predicted death rate vs. the actual average (running average of 100 samples as ranked by the prediction score) death rate for this updated model. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 30.

TABLE 30 Average death rate versus prediction score. Prediction score Number of samples Number of deaths Rate <0.3 59 11 0.186440678 0.3-0.5 32 12 0.375 0.5-0.7 40 24 0.6 0.7-0.9 73 46 0.630136986 >0.9 53 43 0.811320755

Using a threshold of 0.6, the odds ratio for overall survival is 5.7 (95% CI: 3.3-9.9), Fisher's Exact Test p-value=6.7×10−11.

Patients can be further divided into good (risk score<0.4), medium (score 0.4-0.75) and poor (score>0.75) prognosis groups. FIG. 21 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 56.0 (P=6.8×10−13).

Example 6 Prognostic Model for Prostate Cancer

This example describes a prostate cancer prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature was reduced to 10 genes to simplify the implementation of this prognosis model.

A total of 302 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model validated in the second half of samples. In the first half of samples, 151 samples had outcome data (live or death). In the second half of samples, 151 samples had outcome data. The detailed last follow-up dates for the good outcome patients are incomplete. In the first half of samples, 16 out of 137 good outcome patients did not have the last follow-up date. In the second half of samples, 16/127 good outcome patients did not have the last follow-up date. In poor outcome patients, all but one had last follow-up dates.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 151 training samples which were either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 31 & 32. Genes in Table 32 are highly enriched for cell cycle and cell proliferation pathways.

The model was built in the training set using a general linear model (from the R package) using the following equation:


Prostate Cancer Risk Score=0.41973 +0.08610*(prg2−prg1)   (Formula 11),

where “prg1” is a score calculated from prognosis genes in Table 31 and “prg2” is a score calculated from prognosis genes in Table 32. Scores can be calcualted by averaging the log2(intensity) of each probe in the geneset.

The performance of this model is evaluated in reserved validation set of 151 samples.

Using a threshold of 0.4, the odds ratio for overall survival was 51.4 (95% CI: 14.1-186.9), Fisher's Exact Test p-value =2.2×10−11.

The Kaplan-Meier curves using the same threshold are shown in FIG. 22. The Chi-square on 1 degrees of freedom is 123 (P=0).

The number of genes in each pathway was reduced to 10 genes.

Prognosis Signature Component 1 (prg1):

    • Probe IDs: merck-NM_012134_at, merck-NM_021965_s_at, merck-BC064695_s_at, merck2-BF681326_at, merck2-NM_015385_at, merck-NM_032105_at, merck-AF055081_s_at, merck-NM_001299_at, merck2-AI745408_a_at, merck-CA438563_at
    • Gene symbols: LMOD1, PGM5, MYLK, SYNPO2, SORBS1, PPP1R12B, DES, CNN1, MYH11, MYOCD

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck-NM_012112_at, merck-NM_181802_at, merck-NM_004219_x_at, merck2-AK023483_at, merck-NM_001809_at, merck-NM_198436_s_at, merck-NM_080668_at, merck-NM_018454_at, merck-NM_004217_at, merck-ENST00000333706_x_at
    • Gene symbols: TPX2, UBE2C, PTTG1, NUSAP1, CENPA, AURKA, CDCA5, NUSAP1, AURKB, BIRC5,

The scores derived from these 10-genes correlated to the original scores at the level of 0.98 for both prg1 and prg2.

Using the reduced gene sets, the updated predictive model is:


Prosate Cancer Risk Score=0.34044+0.06186*(prg2−prg1)   (Formula 12).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

The performance of the reduced genesets was the same as the original genesets. Using a threshold of 0.4, the odds ratio for overall survival is 51.4 (95% CI: 14.1-186.9), Fisher's Exact Test p-value=2.2×10−11.

The Kaplan-Meier curves using the same threshold are shown in FIG. 23. The Chi-square on 1 degrees of freedom is 123 (P=0).

TABLE 31 Prognosis signature component 1 (anti-correlated with poor outcome) probe Gene merck-NM_021965_s_at PGM5 merck-BC064695_s_at MYLK merck2-NM_152795_at HIF3A PPP5C merck2-BU195365_at LMOD1 merck-NM_005197_s_at FOXN3 merck-NM_032801_at JAM3 merck2-BC036093_at HLF merck-ENST00000343365_a_at LMOD1 merck-AL832580_at RNF180 merck2-BX118828_at merck-NM_001025266_at C3orf70 merck2-AW964876_at FOXN3 merck-NM_004078_at CSRP1 merck-J02854_at MYL9 merck2-AI598275_at CSRP1 merck-AK098218_a_at PGM5-AS1 merck-BQ709647_a_at HLF merck-NM_213674_x_at TPM2 RMRP merck-NM_181526_s_at MYL9 merck-NM_014365_at HSPB8 merck-AK093957_s_at MIR143HG merck2-BX350133_at merck-NM_033303_at ADRA1A merck-NM_003462_at DNALI1 merck-NM_002126_at HLF merck-NM_007177_at FAM107A merck-NM_012134_at LMOD1 merck2-CD557691_at NFIA merck-ENST00000371189_s_at NFIA merck-ENST00000372045_at CHRDL1 merck2-BG674122_a_at HLF merck2-EB387139_a_at ATP1A2 merck2-AI692523_at merck-NM_001042_at SLC2A4 merck2-BF681326_at SYNPO2 merck-NM_013377_at PDZRN4 merck-NM_000898_at MAOB MAOA merck-ENST00000261302_a_at FOXN3 merck2-NM_022844_s_at merck-BC107758_at TNS1 merck-NM_004137_at KCNMB1 KCNIP1 LOC101928033 merck2-NM_015385_at SORBS1 merck-D10667_a_at MYH11 NDE1 merck2-AL532587_at TPM2 RMRP merck2-BC107783_s_at merck-BX381493_s_at ANKRD35 merck-AL833294_s_at SYNPO2 merck2-NM_000195_at HPS1 merck2-AL831991_at ATP1A2 merck2-NM_003734_at AOC3 merck2-DC364710_x_at NEXN merck-ENST00000361490_a_at HPS1 merck-ENST00000330010_a_at NEXN merck-NM_004975_at KCNB1 merck-NM_000961_at PTGIS merck-NM_003734_at AOC3 merck2-AI745408_a_at MYH11 merck2-NM_147162_at IL11RA merck2-BC113456_at MYLK merck2-H40930_at NECAB1 merck-NM_053029_s_at MYLK merck2-CD299407_x_at NEXN merck2-EB387733_a_at SORBS1 merck-BQ888844_a_at SORBS1 merck-ENST00000312358_s_at SPEG merck-AI918006_at UBXN10 merck-NM_002398_at MEIS1 merck-NM_198995_s_at CCDC178 merck2-NM_033254_at merck-BU681386_at SCN7A merck2-CD299407_at NEXN merck-NM_001299_at CNN1 merck-NM_025220_s_at ADAM33 merck-NM_203441_at FRA10AC1 merck2-BX464303_at GSTM3 merck2-ENST00000371953_at PTEN merck-NM_020899_s_at ZBTB4 merck2-H40930_x_at NECAB1 merck-NM_001456_s_at FLNA merck2-NM_001037954_at DIXDC1 merck-AK024986_at PTEN merck2-AL554563_at ACTA2 merck-NM_022062_s_at PKNOX2 merck-AY358229_a_at MSRB3 merck-NM_001387_at DPYSL3 merck2-BC034387_at SLC2A4 merck2-AA536214_at merck-NM_020925_s_at CACHD1 merck-AK056079_s_at JAM2 GABPA merck-AL833622_a_at MSRB3 merck-NM_001083_at PDE5A merck2-BC055084_at NEXN merck2-NM_016826_at OGG1 CAMK1 merck-NM_001759_at CCND2 merck-NM_014057_a_at OGN merck-AK026168_at merck2-AI288607_at merck-NM_145728_at SYNM merck2-AK056845_at merck-NM_002725_at PRELP OPTC

TABLE 32 Prognosis signature component 2 (correlated with poor outcome) probe Gene merck2-AF225416_at SPC25 merck-NM_020675_at SPC25 merck-BC003664_a_at KIF4A merck2-NM_024037_at AUNIP merck-NM_001809_at CENPA merck-NM_181802_at UBE2C merck-NM_014176_at UBE2T merck-NM_005733_at KIF20A CDC23 merck-NM_013277_a_at RACGAP1 merck-CR602847_a_at KIAA0101 merck2-DQ890621_at CDC45 merck-NM_018248_at NEIL3 merck-BC035392_at HMMR merck2-NM_005196_at CENPF merck-NM_004219_x_at PTTG1 merck2-AK097710_at CDC25C merck-NM_001786_a_at CDK1 RHOBTB1 merck-NM_144508_at CASC5 merck-NM_016343_at CENPF merck-DA823877_a_at CDK1 RHOBTB1 merck-NM_152259_s_at TICRR KIF7 merck-NM_004701_at CCNB2 merck-NM_003504_at CDC45 merck-AK055176_s_at FANCI merck-BC075828_a_at GTSE1 merck-NM_203394_at E2F7 merck-NM_001039841_s_at ARHGAP11A ARHGAP11B merck-NM_001790_at CDC25C merck-NM_004217_at AURKB merck-NM_002497_at NEK2 merck-ENST00000246083_s_at DNAJC9 ZFYVE26 merck2-AB046790_at CASC5 merck-NM_031299_at CDCA3 GNB3 merck-BC048988_a_at SKA3 merck-NM_016426_at GTSE1 TRMU merck-NM_014750_at DLGAP5 merck-NM_021953_at FOXM1 merck2-BC107750_at CDK1 RHOBTB1 merck-NM_014791_at MELK merck-NM_002466_at MYBL2 merck-NM_001067_at TOP2A merck2-NM_203399_at STMN1 merck-NM_130398_at EXO1 merck-NM_006461_at SPAG5 merck2-BX091454_a_at RACGAP1 merck2-BE856617_at AURKA merck-NM_080668_at CDCA5 merck-AK093235_s_at TDP1 merck2-AF043294_at BUB1 RGPD6 merck2-DB485269_a_at merck-NM_018101_at CDCA8 merck-BC024211_a_at NCAPH merck-NM_012310_at KIF4A GDPD2 merck-NM_018136_s_at ASPM merck-BF511624_s_at BUB1B merck-NM_012112_at TPX2 merck2-ENST00000372927_at CENPI merck2-BC006325_x_at GTSE1 TRMU merck-AK129748_s_at STMN1 merck-BF308644_s_at CENPI merck-NM_174942_a_at GAS2L3 merck-NM_198436_s_at AURKA merck-NM_002417_at MKI67 merck-NM_001255_s_at CDC20 merck2-AK025810_at WDR5 merck-NM_003258_at TK1 merck2-DQ892840_a_at CDC6 merck-NM_003201_at TFAM merck-NM_017669_at ERCC6L merck2-BC014353_a_at STMN1 merck-CR622584_s_at CHEK2 merck-NM_004336_at BUB1 RGPD6 merck2-AL517462_s_at merck-AK057037_at FEZF1-AS1 merck2-AL703195_s_at merck-NM_001002876_at CENPM merck-NM_004203_a_at PKMYT1 merck2-XM_937756_a_at FEN1 merck-ENST00000243201_a_at HJURP merck-ENST00000373940_a_at ZWINT merck-AI418253_at PMS2LP2 merck-BI868409_a_at MKI67 merck2-ENST00000373899_at TFAM merck-NM_020394_at ZNF695 ZNF670-ZNF695 merck-BQ653044_a_at EZH2 merck-CR602926_s_at CCNB1 merck2-NM_018944_at MIS18A merck-NM_032117_at MND1 merck-NM_018454_at NUSAP1 merck-NM_005192_at CDKN3 merck-BC038772_s_at MCM4 merck2-BT006759_at KIF2C merck-CR596700_a_at RRM2 merck2-BC106011_a_at ACP1 merck2-AK023483_at NUSAP1 merck-NM_003533_at HIST1H3I merck2-BC022400_at METTL6 merck2-BC034607_at ASPM merck2-NM_031966_at CCNB1 merck-NM_138419_s_at MTFR2

Example 7 Prognostic Model for Pancreatic Cancer

This example describes a pancreatic cancer prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

A total of 525 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model validated using the second half of samples. In the first half of samples, 261 samples had outcome data (live or death). In the second half of samples, also 263 samples had outcome data. The detailed last follow-up dates for the good outcome patients are incomplete. In the first half of samples, 12 out of 97 good outcome patients did not have the last follow-up date. In the second half of samples, 30/136 good outcome patients did not have the last follow-up date.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 261 training samples which are either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 33 & 34. Genes in Table 34 are highly enriched for cell cycle and cell proliferation pathways.

A model was built in the training set using a general linear model (from the R package) using the following equation:


Pancreatic Cancer Risk Score=Risk Score=0.467962 +0.076686*(prg2−prg1)   (Formula 13),

where “prg1” is a score calculated from prognosis genes in Table 33 and “prg2” is a score calculated from prognosis genes in Table 34. The scores can be calculated by averaging the log2(intensity) of each probe in the geneset.

The performance of this model is evaluated in reserved validation set of 263 samples.

Using a threshold of 0.5, the odds ratio for overall survival was 35.2 (95% CI:6 8.3-148), Fisher's Exact Test p-value=3.7×10−14.

The Kaplan-Meier curves using the same threshold is shown in FIG. 24. The Chi-square on 1 degrees of freedom is 33.9 (P=5.82×10−9).

The number of genes in each pathway was reduced to 10 genes.

Prognosis Signature Component 1 (prg1):

    • Probe IDs: merck2-AL133657_at, merck2-NM_033026_at, merck-NM_018711_at, merck-BC001946_a_at, merck-NM_006650_at, merck-BI552493_a_at, merck-ENST00000371069_a_at, merck-NM_004644_at, merck-BC045704 a_at,merck2-NM_005374_at
    • Gene symbols: RUNDC3A, PCLO, SVOP, CELF4, CPLX2, SCG3, DNAJC6, AP3B2, SCN3B, MPP2

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck-NM_006142_at, merck-NM_000228_at, merck2-NM_183247_a_at, merck-NM_016445_at, merck-NM_002447_at, merck-NM024009_at merck-NM_080388 at merck-NM_003979 at merck-NM_001005376 at merck-NM_001747_at
    • Gene symbols: SFN, LAMB3, TMPRSS4, PLEK2, MST1R, GJB3, S100A16, GPRC5A, PLAUR, CAPG

The scores derived from these 10-genes correlated to the original scores at the level of 0.97 for prg1 and 0.98 for prg2.

Using the reduced gene sets, the updated predictive model is:


Pancreatic Cancer Risk Score=Risk Score=0.504576+0.049284*(prg2−prg1)   (Formula 14).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

The performance of the reduced genesets is similar the original genesets. Using a threshold of 0.5, the odds ratio for overall survival is 22.5 (95% CI: 6.8-74.7), Fisher's Exact Test p-value=8.4×10−13. The Kaplan-Meier curves using the same threshold are shown in FIG. 25. The Chi-square on 1 degrees of freedom is 30.2 (P=3.8×10−8).

TABLE 33 Prognosis signature component 1 (anti-correlated with poor outcome) probe Gene merck-NM_024557_at RIC3 merck-NM_171998_at RAB39B merck-ENST00000379272_at ACSL6 merck-XM_938173_at CELF4 merck-NM_024026_x_at MRP63 merck-BC001946_a_at CELF4 merck2-BX647514_a_at RIC3 merck2-NM_020180_at CELF4 merck2-DB523436_at ACSL6 merck-AK056249_at merck2-AL832601_at RIC3 TUB merck-NM_144576_at COQ10A merck-NM_020818_at UNC79 merck2-AL133657_at RUNDC3A merck-AK075495_at NDFIP1 merck-NM_030802_at FAM117A merck-BC044777_at TMX4 merck-NM_006695_a_at RUNDC3A merck-NM_032829_at FAM222A merck2-AL532654_at CIRBP merck-AK125327_a_at UNC79 merck-BG212691_s_at EPM2A merck-ENST00000377770_a_at DPP6 merck2-NM_138362_at FAM104B merck-CR605402_at TBCK merck2-AF546872_at PACRG merck-NM_020708_at SLC12A5 merck-AW297465_at merck2-BI761148_a_at CIRBP merck2-AK092094_at SLC25A5-AS1 SLC25A5 merck-NM_152410_at PACRG merck-BC037882_at merck-NM_020949_s_at SLC7A14 merck-AK055712_at LOC728705 merck-NM_022151_at MOAP1 merck-NM_138362_at FAM104B merck-NM_003179_at SYP PRICKLE3 merck-NM_021156_a_at TMX4 merck-NM_006650_at CPLX2 merck-NM_001033002_s_at RPAIN merck-NM_170710_at WDR17 merck2-NM_033026_at PCLO merck-BU170673_at merck-NM_016188_at ACTL6B TFR2 merck2-BC028357_at CLGN merck2-AL832187_at ARMCX5-GPRASP2 GPRASP2 BHLHB9 merck-NM_001280_a_at CIRBP merck-BX640845_a_at FSTL4 merck2-AK094546_at QDPR merck2-NM_172232_at ABCA5 merck2-ENST00000379240_at ACSL6 merck-NM_004362_at CLGN merck-NM_001039350_at DPP6 merck-BC035377_at DMTF1 merck-AF052119_at SLC25A4 merck2-AK074845_x_at NUDT9 merck2-AK093871_at CXXC4 merck-ENST00000332709_at PGRMC2 merck-BC018917_a_at MYT1 merck-BC009714_a_at RAB39B merck-CA868555_a_at RIC3 merck-NM_007185_at CELF3 merck-AK094547_at SLC7A14 merck2-BM977387_at merck-ENST00000371069_a_at DNAJC6 merck-NM_144611_s_at CYB5D2 merck2-DB479534_at BEX2 merck2-BY798024_at UNC80 merck-NM_173092_a_at KCNH6 DCAF7 merck-AI474150_a_at ISCA1 merck2-BU687744_at merck-NM_152503_at MROH8 merck2-CK903584_at SERPINI1 merck-NM_019114_at EPB41L4B merck-NM_014723_at SNPH SDCBP2 merck2-CD742622_at TARBP2 merck-CK819476_s_at XPNPEP2 merck-AF086195_at DCUN1D5 merck-NM_145170_at TTC18 merck2-BC020263_at CYB5D2 merck2-NM_019589_at YLPM1 merck2-BF224377_at merck-CR596771_a_at QDPR merck-AK123831_at CDS2 merck2-BF433548_at merck-NM_015063_at SLC8A2 merck-NM_025212_a_at CXXC4 LOC101929468 merck-BX537526_at SLC24A5 merck2-BG695979_at merck-AK090762_s_at merck2-AL517382_at AKAP14 merck-AK127804_at RFX3 LOC101929247 merck-AK123201_at MTMR7 VPS37A merck-BM681832_at merck-AK127501_at merck-AK002023_at CTDP1 merck-NM_033053_s_at DMRTC1 DMRTC1B merck-AK124803_at PGBD5 merck2-BF304197_at merck-ENST00000372943_at FITM2

TABLE 34 Prognosis signature component 2 (correlated with poor outcome) probe Gene merck-NM_001747_at CAPG merck-NM_004004_s_at GJB2 merck2-BC071703_at GJB2 merck-NM_006142_at SFN merck2-AF177862_a_at HN1 merck-NM_000228_at LAMB3 merck-NM_080388_at S100A16 merck-NM_007267_at TMC6 merck2-NM_009587_s_at merck-NM_018685_at ANLN merck2-NM_001048201_at UHRF1 merck2-NM_001042685_s_at merck2-CR936650_at ANLN merck2-X74039_at PLAUR merck-NM_001005376_at PLAUR merck-NM_000213_at ITGB4 GALK1 merck2-AF491781_a_at OSBPL3 merck-NM_018131_at CEP55 merck-BC017731_a_at OSBPL3 merck-BC105943_s_at LGALS9 LGALS9B LGALS9C FAM106B merck2-NM_001042422_at SLC16A3 merck-NM_003979_at GPRC5A merck-NM_006681_at NMU merck2-BM543893_x_at PLAUR merck-NM_005980_at S100P merck-X15014_a_at RALA merck2-AF318350_at TTYH3 merck2-BG680883_at merck-BC046920_a_at NQO1 merck-CR407664_a_at PHLDA2 merck-BI868409_a_at MKI67 merck2-AK223027_at PHLDA2 merck-BG677853_a_at LAMC2 merck-NM_005620_at S100A11 merck2-NM_183247_a_at TMPRSS4 merck-AF086216_at SERPINB5 merck-NM_005562_at LAMC2 merck-NM_145903_s_at HMGA1 merck2-NM_001005377_at PLAUR merck2-AK097588_at ATL3 merck-NM_018715_a_at RCC2 merck-NM_000189_at HK2 merck-NM_001005377_s_at PLAUR merck-NM_019034_at RHOF TMEM120B merck-AI924527_a_at TMPRSS4 merck-BC042436_at merck-NM_015459_s_at ATL3 merck-BM806310_a_at OSBPL3 merck2-BC013892_at PVRL4 merck-NM_001037330_s_at TRIM16L TRIM16 merck2-AL517462_s_at merck-CR596700_a_at RRM2 merck-NM_014568_s_at GALNT5 merck-NM_025250_at TTYH3 merck2-AI701192_at LAMC2 merck-NM_002639_at SERPINB5 merck-NM_004701_at CCNB2 merck-NM_012112_at TPX2 merck-NM_001793_at CDH3 merck2-BG675923_x_at merck2-AI701192_x_at LAMC2 merck2-AV714642_at ANLN merck-NM_002447_at MST1R merck-NM_033520_at C19orf33 YIF1B PPP1R14A merck-NM_014791_at MELK merck2-M62898_x_at ANXA2 merck-NM_000422_x_at KRT17 merck-NM_000445_at PLEC merck-ENST00000335534_s_at KIF18B merck-NM_002250_at KCNN4 merck2-AF098158_at TPX2 merck-NM_014624_at S100A6 merck-CR607300_a_at MKI67 merck-NM_003844_at TNFRSF10A merck-NM_181802_at UBE2C merck-NM_002068_at GNA15 merck-BC001459_s_at RAD51 merck-NM_005975_at PTK6 merck-AY358204_a_at TMEM92 merck2-AF070544_at SLC2A1 merck2-NM_001083947_at TMPRSS4 merck-NM_012101_at TRIM29 merck2-AL831846_at CELSR1 merck-NM_002417_at MKI67 merck-AL582254_x_at merck2-NM_005975_a_at merck2-BT009912_x_at merck-AB208913_a_at ITGB4 merck-NM_014750_at DLGAP5 merck2-BT009912_at merck-NM_003258_at TK1 merck-NM_024009_at GJB3 merck-NM_199129_at TMEM189 merck-NM_016445_at PLEK2 merck-NM_002306_s_at LGALS3 merck-NM_021103_a_at TMSB10 merck-NM_005978_at S100A2 merck-NM_020672_at S100A14 merck-ENST00000360566_at RRM2 merck-NM_025049_at PIF1

Example 8 Prognostic Model for Endometrium Cancer

This example describes an endometrium cancer prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

A total of 410 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model validated using the second half of samples. In the first half of samples, 204 samples had outcome data (alive or dead). Among them, 140 had good outcome and 64 had poor outcome. In the good outcome patients, 12 did not have tumor grade data, and in the poor outcome patients, 17 did not have tumor grade data. In the second half of samples, also 204 had outcome data. Among them, 158 had good outcome and 46 had poor outcome. 13 and 7 patients did not have tumor grade data in good and poor outcome patients respectively.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 204 training samples which are either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 35 & 36. Genes in Table 36 are highly enriched for cell cycle and cell proliferation pathways.

A model was built in the training set using a general linear model (from the R package) using the following equation:


Endometrium Cancer Risk Score=Risk Score=0.01786 +0.08208*(prg2−prg1)+(0.14297*Grade)   (Formula 15),

where “prg1” is a score calculated from prognosis genes in Table 35 and “prg2” is a score calculated from prognosis genes in Table 36. The scores can be calculated by averaging the log2(intensity) of each probe in the geneset. It's worth pointing out that PGR, ESR1 and AR are all in Table 35, and Table 36 is enriched for proliferation genes. Grade represents tumor grade.

The performance of this model is evaluated in reserved validation set of 184 samples with both gene expression and tumor grade data. FIG. 26 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 37.

TABLE 37 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.1 67 9 0.134 0.1-0.3 63 11 0.175 0.3-0.5 33 8 0.242 >0.5 21 11 0.524

Using a threshold of 0.2, the odds ratio for overall survival is 3.8 (95% CI: 1.8-8.1), Fisher's Exact Test p-value=4.8×10−4.

Patients can be further divided into good (risk score<0.2), medium (score 0.2-0.4) and poor (score>0.4) prognosis groups. FIG. 27 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 18.5 (P=9.7×10−5).

The number of genes in each pathway was reduced to 10 genes.

Prognosis Signature Component 1 (prg1):

    • Probe IDs: merck-AF016381_a_at, merck-AI918006_at, merck2-NM_001080537_at, merck-NM_145263_at, merck2-NM_173615_at, merck2-XM_371638_at, merck-NM_025145_at, merck2-NM_016930_at, merck-NM_173081_at, merck-AL040975_at
    • Gene symbols: PGR, UBXN10, SNTN, SPATA18, VWA3A, CDHR4, WDR96, STX18, ARMC3, ESR1

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck2-BM904739_at, merck-ENST00000311926_s_at, merck-NM_003875_at, merck-NM_007274_s_at, merck-NM_005225_at, merck-AK027859_s_at, merck-NM_018270_at, merck-NM_198436_s_at, merck2-NM_001168_at, merck2-AF098158_at
    • Gene symbols: MRGBP, UBE2S, GMPS, ACOT7, E2F1, CENPO, MRGBP, AURKA, BIRC5, TPX2

The scores derived from these 10-genes are correlated to the original scores at the level of 0.96 for prg1, 0.85 for prg2.

Using the reduced gene sets, the updated predictive model is:


Endometrium Cancer Risk Score=Risk Score=−0.13842+0.04180*(prg2−prg1)+(0.18547*Grade)   (Formula 16).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

In the validation set, patients are grouped by the prediction score. Table 38 shows the detailed information about number of samples, number of deaths, and the death rate in each prediction score bin.

TABLE 38 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.2 89 10 0.112 0.2-0.4 53 12 0.226 0.4-0.6 36 13 0.361 >0.6 6 4 0.667

Using a threshold of 0.2, the odds ratio for overall survival is 3.5 (95% CI: 1.6-7.6), Fisher's Exact Test p-value=2.1×10−3.

Patients can be further divided into good (risk score<0.2), medium (score 0.2-0.4) and poor (score>0.4) prognosis groups. FIG. 28 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 18.4 (P=1.0×10−4).

TABLE 35 Prognosis signature component 1 (anti-correlated with poor outcome) probe Gene Hmerck-BX106921_at PGR merck-AL137566_at PGR merck-AF016381_a_at PGR merck-AL040975_at ESR1 merck-ENST00000369936_at KIAA1324 merck2-AL050116_at ESR1 merck-BX647987_at LOC100507053 merck-AL702564_at PGR merck2-NM_000125_at ESR1 merck-NM_000125_at ESR1 merck-AI918006_at UBXN10 merck2-BX648631_at UBXN10 merck2-NM_016930_at STX18 merck-NM_145263_at SPATA18 merck-NM_001025593_at ARFIP1 merck-AW970795_at merck-NM_152376_s_at UBXN10 merck2-AI288607_at merck2-M69297_at merck-NM_020775_s_at KIAA1324 merck2-BM695584_at ARHGAP26 merck2-NM_006961_at ZNF19 merck-NM_013367_s_at ANAPC4 merck-NM_000266_at NDP merck-NM_025059_at CCDC170 merck-CR609491_a_at STX18 merck2-NM_005327_at HADH merck-ENST00000324607_s_at MBOAT1 merck2-CA309763_at NDP merck-ENST00000369949_s_at C1orf194 merck-NM_014668_s_at GREB1 merck-NM_025145_at WDR96 merck-NM_001002912_s_at C1orf173 merck2-ENST00000342217_at C1orf173 merck2-AK025905_at SOX17 merck-BC094795_a_at PIK3R1 merck2-BG619802_at EYA2 merck-NM_015071_at ARHGAP26 merck-BX648957_at LOC100505776 merck-BC028018_at LOC100129098 merck-NM_178456_at C20orf85 merck-NM_022454_at SOX17 merck-ENST00000347491_s_at ESR1 merck-NM_214462_at DACT2 merck-NM_003551_at NME5 merck-ENST00000319471_a_at SORBS2 merck2-AM392558_at SORBS2 merck2-CB999963_at RNF180 merck-NM_181523_at PIK3R1 merck-NM_018242_at SLC47A1 merck-AK057330_a_at ZNF19 merck-NM_022123_a_at NPAS3 merck2-BQ894504_at PIK3R1 merck-BC063677_at TMEM231 CHST5 merck-NM_145170_at TTC18 merck-BC063866_at COL28A1 merck-NM_003774_at POC1B-GALNT4 GALNT4 merck-NM_018043_at ANO1 merck2-AY358612_at TMEM231 CHST5 merck-AF085947_at NPAS3 merck-NM_015460_at MYRIP merck2-DT217746_at ASRGL1 merck2-AK225360_at SLC47A1 merck2-NM_001080537_at SNTN merck-CF453637_s_at NPAS3 merck2-BX093691_at TTC18 merck-NM_004816_s_at FAM189A2 merck-ENST00000299840_s_at VWA3A merck-BC037328_at MAP2K6 merck-AL832580_at RNF180 merck2-NM_144722_at SPEF2 merck-NM_005244_at EYA2 merck-NM_025080_s_at ASRGL1 merck-AI624058_at FAM216B merck2-ENST00000374690_at AR merck-NM_018091_s_at ELP3 merck-XM_942673_at SNTN merck2-BX648791_at merck-CD687039_a_at DNAH12 merck2-BQ684833_at ACSL5 merck2-BX096668_at merck-AY312852_s_at GTF2IRD2 GTF2IRD2B GTF2I merck-NM_145058_at RILPL2 merck-NM_201520_s_at SLC25A35 RANGRF merck-BC047078_at SLC25A15 merck2-NM_173615_at VWA3A merck-NM_015058_at VWA8 merck2-NM_173537_s_at merck2-NM_001003795_s_at merck-T68445_a_at AR merck2-XM_371638_at CDHR4 merck2-BC026182_at NME5 merck-NM_005397_at PODXL MKLN1 merck-NM_001029875_at RGS7BP merck-NM_015271_at TRIM2 merck2-BC047091_a_at ZNF19 merck2-AA148029_at PODXL MKLN1 merck2-NM_145283_at NXNL2 merck-AL050026_at PALLD merck-NM_020879_s_at CCDC146

TABLE 36 Prognosis signature component 2 (correlated with poor outcome) probe Gene merck2-BM904739_at MRGBP merck-NM_018270_at MRGBP merck-NM_007274_s_at ACOT7 merck-NM_004358_at CDC25B merck2-BQ437524_at CDC25B merck-AF533230_x_at USP32 merck2-BX647988_a_at CDC25B merck2-BC007074_a_at TNNT1 merck2-BC001395_at CIAO1 merck2-ENST00000356433_at DLL3 merck-BX442394_a_at SOX11 merck2-BQ644821_at merck2-AK026140_at merck-XM_926989_s_at ACAA2 merck-CR609746_a_at C17orf96 merck-NM_138570_s_at SLC38A10 merck-NM_001010911_at CASC10 merck2-AY762903_at TNNT1 merck-NM_003283_s_at TNNT1 merck2-DQ893376_s_at ACAA2 merck2-BC002615_at CSNK2A1 CSNK2A3 merck-NM_001031713_s_at MCUR1 merck-BC003580_s_at CIAO1 merck-NM_003108_at SOX11 merck-NM_021972_at SPHK1 merck2-DQ893376_at ACAA2 merck-NM_004181_at UCHL1 merck-BC037270_a_at AKAP8 merck-NM_001039467_s_at RGS19 merck-NM_203486_s_at DLL3 merck-NM_153485_at NUP155 merck-ENST00000311926_s_at UBE2S merck-NM_006111_at ACAA2 merck-NM_004708_s_at PDCD5 merck-NM_021158_at TRIB3 merck-ENST00000381973_s_at CSNK2A1 CSNK2A3 merck-NM_000071_s_at CBS U2AF1 merck-NM_004209_at SYNGR3 merck-NM_152310_at ELOVL3 PITX3 merck-NM_004112_at FGF11 CHRNB1 merck2-BI602361_s_at merck2-BC068553_at DR1 merck-DW451489_s_at MED8 merck-NM_002808_at PSMD2 merck-CR610223_a_at SCARB2 merck-NM_003875_at GMPS merck-BC028386_a_at RRP1B merck-CR619305_a_at GNB1 merck-NM_000022_at ADA merck-CR592459_a_at MAPRE1 merck2-BC030582_at TCP11L1 merck2-BC002615_s_at CSNK2A1 CSNK2A3 merck-NM_001089_at ABCA3 merck-NM_015122_at FCHO1 merck-NM_001281_at TBCB merck-NM_001489_a_at NR6A1 merck-AK023842_a_at BAZ2A merck-NM_002792_s_at PSMA7 merck-BC025264_a_at YTHDF1 merck-NM_001426_at EN1 merck-NM_003198_at TCEB3 merck2-ENST00000305989_at FTL GYS1 merck-AK027859_s_at CENPO merck-ENST00000264607_a_at ASB1 merck-NM_013409_at FST merck-NM_080618_at CTCFL merck2-BQ227259_at SCARB2 merck-BX649059_at GAS2L3 merck-NM_152699_s_at SENP5 merck-NM_014109_a_at ATAD2 merck-AK126101_a_at PLXNA1 merck-NM_004341_at CAD merck2-NM_001079862_at DBI merck-NM_013321_at SNX8 merck2-EF560732_a_at CKAP2 merck-CR617826_a_at TIMM50 merck2-BC007338_at CDV3 merck-NM_206831_a_at DPH3 OXNAD1 RFTN1 merck2-ENST00000374536_at TCEB3 merck-NM_007224_at NXPH4 SHMT2 merck-ENST00000373683_s_at SKA2 merck2-AA169659_s_at merck2-BC121146_at TIMM50 merck2-ENST00000305989_x_at FTL GYS1 merck-BM722157_a_at SOX11 merck-BM909568_s_at PRMT2 S100B merck2-BC025843_at L1CAM merck-NM_024871_at MAP6D1 merck2-BE264170_at PLCXD1 merck-NM_003088_at FSCN1 merck2-AK025810_at WDR5 merck2-BM674474_at merck-BU145850_at merck2-AK222554_at SF3A3 merck2-AF225416_at SPC25 merck-NM_198207_at CERS1 merck2-AI149996_at ADRM1 merck-NM_000175_s_at GPI merck-AK074937_a_at NETO2 merck-ENST00000330234_a_at DGCR5

Example 9 Prognostic Model for Melanoma

This example describes a melanoma prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

A total of 711 samples were profiled by Affymetrix® expression arrays, of which 559 were malignant melanoma. A composite model was built using the first half of samples and the model validated using the second half of samples. In the first half of samples, 292 samples had outcome data (alive or dead). Among them, 123 had good outcome and 169 had poor outcome. In the second half of samples, all 267 had outcome data. Among them, 105 had good outcome and 162 had poor outcome. Besides malignant melanoma, there are also 152 other skin cancer samples including squamous cell carcinoma, Merkel cell carcinoma, Basal cell carcinoma, etc. The model developed by malignant melanoma was also evaluated in these 152 samples.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 267 training samples which are either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 37 & 38. Genes in Table 38 are highly enriched for cell cycle and cell proliferation pathways.

A model was built in the training set using a general linear model (from the R package) using the following equation:


Melanoma Cancer Risk Score=Risk Score=0.16708+0.10739*(prg2−prg1)   (Formula 17),

where “prg1” is a score calculated from prognosis genes in Table 37 and “prg2” is a score calculated from prognosis genes in Table 38. The scores can be calculated by averaging the log2(intensity) of each probe in the geneset.

The performance of this model is evaluated in reserved validation set of 267 samples with also the stage data. FIG. 29 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 38.

TABLE 38 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.4 45 18 0.400 0.4-0.5 32 15 0.469 0.5-0.6 47 24 0.511 0.6-0.7 66 49 0.742 >0.7 77 56 0.727

Using a threshold of 0.58, the odds ratio for overall survival is 3.0, 95% CI: 1.8-5.0, Fisher's Exact Test p-value=2.5×10−5.

Patients can be further divided into good (risk score<0.45), medium (score 0.45-0.65) and poor (score>0.65) prognosis groups. FIG. 30 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 37.0 (P=9.3×10−9).

The number of genes in each pathway was reduced to 10 genes.

Prognosis Signature Component 1 (prg1):

    • Probe IDs: merck-AK128436_at, merck-NM_000073_at, merck-NM_002351_s_at, merck2-NM_052931_at, merck-NM_000734_at, merck-NM_052931_at, merck-NM_018556_s_at, merck2-NM_025228_at, merck2-NM_001010923_at, merck-NM_198517_at
    • Gene symbols: IKZF3, CD3G, SH2D1A, SLAMF6, CD247, SLAMF6, SIRPG, TRAF3IP3, THEMIS, TBC1D10C

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck-NM_032039_at, merck-NM_001010866_at, merck2-AL157485_at, merck-ENST00000336690_s_at, merck-NM_014291_at, merck-NM_001014832_s_at, merck-BM981759_a_at, merck-ENST00000372943_at, merck-ENST00000360797_s_at, merck2-CA311625_at
    • Gene symbols: ITFG3, TMEM201, TBC1D16, PPT2, GCAT, PAK4, OTUD7B, FITM2, PCGF2, GCAT

The scores derived from these 10-genes are correlated to the original scores at the level of 0.98 for prg1, 0.87 for prg2.

Using the reduced gene sets, the updated predictive model is:


Melanoma Cancer Risk Score=Risk Score=0.43492+0.06120*(prg2−prg1)   (Formula 18).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

FIG. 31 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate for this updated model. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 39.

TABLE 39 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.4 36 14 0.389 0.4-0.5 46 24 0.522 0.5-0.6 66 34 0.515 0.6-0.7 69 53 0.768 >0.7 50 37 0.740

Using a threshold of 0.6, the odds ratio for overall survival is 3.3 (95% CI: 1.9-5.6), Fisher's Exact Test p-value=8.9×10−6.

Patients can be further divided into good (risk score<0.45), medium (score 0.45-0.6) and poor (score>0.6) prognosis groups. FIG. 32 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 32.2 (P=1.0×10−7).

The Model is predictive in other skin cancers: Besides malignant melanoma, there are also 152 other skin cancer samples including squamous cell carcinoma, Merkel cell carcinoma, Basal cell carcinoma, etc. The same model was applied to these 152 samples to evaluate its predictive power.

At a threshold of 0.45, the odds ratio is 5.4, 95% CI: 1.9-15.1, Fisher's exact P-value is 6.3×10−4.

FIG. 33 shows the Kaplan-Meier curves when patients are divided into 3 groups (<0.45, 0.45-0.6 and >0.6). The Chi-square for 2 degrees of freedom is 14 (P=9.2×10−4).

TABLE 37 Prognosis signature component 1 (anti-correlated with poor outcome) probe Gene merck-AI912585_at merck-AK124031_a_at THEMIS merck-NM_016388_at TRAT1 merck2-AY292266_at merck-NM_173799_at TIGIT merck-NM_000619_at IFNG merck-NM_002351_s_at SH2D1A merck-NM_001001895_at UBASH3A merck-NM_012092_at ICOS merck-ENST00000383671_a_at TIGIT merck2-ENST00000390352_x_at merck-Z22965_s_at merck2-NM_004931_a_at CD8B merck-BC036924_at PATL2 SPG11 merck-NM_000073_at CD3G merck2-U39114_s_at merck-NM_198333_s_at P2RY10 merck-DT807100_at CD3D CD3G merck2-AY292266_x_at merck2-BX108263_at LOC101929510 LOC101929531 merck2-ENST00000390435_x_at TRAV8-3 MGC40069 merck-NM_013308_at GPR171 merck-BX648371_at LINC00861 merck2-NM_001010923_at THEMIS merck-ENST00000206681_at merck2-NM_152615_at PARP15 merck-Z75948_s_at TRAV14DV4 merck-CD700761_s_at PPP1R16B merck2-ENST00000390353_at IFI6 TRBV6-1 merck2-ENST00000390352_at merck2-ENST00000390400_at TRBV28 merck2-BM677447_at MIAT merck-NM_172101_at CD8B merck-NM_152693_a_at FAM226A FAM226B merck-AK124004_at AKAP5 merck2-AF459027_at FCRL3 merck-NM_003151_a_at STAT4 merck2-AY006176_x_at merck2-AW170566_at merck2-ENST00000390386_a_at TRBV12-3 TRBV12-4 merck2-ENST00000390363_at merck-CR597260_at LOC101059954 merck-AK097158_at LINC00996 merck2-ENST00000390454_at merck-ENST00000341173_s_at TRAF3IP3 merck2-NM_025228_at TRAF3IP3 merck-NM_032553_at GPR174 merck2-X92770_x_at merck-BC040064_at ITGB2-AS1 ITGB2 merck-ENST00000316577_s_at TESPA1 merck2-ENST00000390439_at merck2-AJ007770_at merck-NM_014450_at SIT1 RMRP merck-AK127925_at CD2 merck-ENST00000303432_a_at CD8B merck2-ENST00000390387_a_at TRBV12-3 TRBV12-4 merck2-AF532855_x_at merck2-ENST00000390435_at TRAV8-3 MGC40069 merck2-ENST00000390449_at merck2-ENST00000390350_at merck2-ENST00000390433_at merck2-ENST00000390393_at TRBV19 merck-Y15200_s_at merck-AK098833_s_at MIAT merck-AY190088_s_at merck-AI281804_at GPR174 merck2-M27337_x_at TRGV2 TRGV4 merck2-L01087_at PRKCQ merck-AF327297_s_at TRAJ17 merck-AK128436_at IKZF3 merck2-ENST00000390394_s_at merck2-ENST00000390359_x_at TRBV4-2 TRBV7-2 merck2-Z22966_a_at merck-NM_005292_at GPR18 merck2-NM_001006638_at RAB37 SLC9A3R1 merck-NM_002262_at KLRD1 merck-NM_152781_at C17orf66 merck-NM_000732_at CD3D merck-NM_000639_at FASLG merck-NM_153615_s_at RGL4 merck2-ENST00000390359_at TRBV4-2 TRBV7-2 merck2-AJ007771_at TRAV8-6 merck-NM_014716_at ACAP1 merck-NM_032206_a_at NLRC5 merck-NM_001024667_s_at FCRL3 merck-NM_198517_at TBC1D10C merck2-ENST00000390353_x_at IFI6 TRBV6-1 merck-NM_000595_a_at LTA merck-BF870822_at merck-ENST00000379833_at GVINP1 merck2-ENST00000390442_at TRAV12-3 merck2-AF129512_at IKZF3 merck-NM_006566_at CD226 merck-AK095686_s_at MIAT merck-BC028218_a_at ZBP1 merck-NM_006257_at PRKCQ merck-NM_018556_s_at SIRPG merck-AI203370_at GBP5 merck2-NM_001005176_a_at SP140 merck-BM700951_at KLRK1 KLRC4-KLRK1

TABLE 38 Prognosis signature component 2 (correlated with poor outcome) probe Gene merck-NM_005027_s_at PIK3R2 merck-NM_001015055_s_at RTKN merck2-BT019930_a_at merck2-BC001528_at merck2-NM_178121_at MEGF8 merck2-NM_003250_a_at THRA NR1D1 merck-NM_178148_at SLC35B2 HSP90AB1 merck-NM_178121_at MEGF8 merck-NM_181521_at CMTM4 merck-CR619245_a_at BSG merck2-AB018267_at IPO13 merck-AK222827_a_at GGCX merck2-BM464059_at merck2-NM_198591_at BSG merck-H05603_a_at THRA NR1D1 merck2-NM_001078172_at FAM127B merck-AF086201_at TMEM63B merck-NM_032039_at ITFG3 merck-NM_003872_s_at NRP2 merck-NM_004793_s_at LONP1 RPL36 merck-ENST00000375101_a_at AGPAT1 merck-NM_018426_at TMEM63B merck-NM_001069_at TUBB2A merck-NM_032806_at POMGNT2 merck-NM_003051_at SLC16A1 merck-AK128554_at IRGQ merck2-CX758384_at DDR1 merck-NM_024085_at ATG9A ABCB6 merck-NM_032088_s_at PCDHGA1 PCDHGA10 PCDHGA11 PCDHGA12 PCDHGA2 PCDHGA3 PCDHGA4 PCDHGA5 PCDHGA6 PCDHGA7 PCDHGA8 PCDHGA9 PCDHGB1 PCDHGB2 PCDHGB3 PCDHGB4 PCDHGB5 PCDHGB6 PCDHGB7 PCDHGC3 PCDHGC4 PCDHGC5 merck-NM_001954_a_at DDR1 merck-NM_015388_s_at YIPF3 merck-NM_014623_at MEA1 merck-ENST00000372943_at FITM2 merck-NM_004053_at BYSL merck-NM_018028_at SAMD4B merck-NM_001012981_at ZKSCAN2 merck-ENST00000321333_x_at FAM127B merck2-BU553968_x_at merck2-NM_000821_at GGCX merck-NM_006876_at B3GNT1 merck-ENST00000261497_at USP22 merck-ENST00000372235_a_at TMEM53 merck2-BC016713_a_at PARVA merck-BC001048_s_at CDK16 merck2-NM_003250_at merck-ENST00000263381_a_at WIZ merck-ENST00000336690_s_at PPT2 merck-NM_001410_at MEGF8 merck-NM_004854_at CHST10 merck-ENST00000360797_s_at PCGF2 merck-AI263624_a_at POFUT1 merck-NM_001035507_a_at AGBL5 merck-NM_001024736_s_at CD276 merck-CR624090_a_at PARVA merck-NM_004860_at FXR2 merck2-AK055481_at SAE1 merck2-BI093105_at NR1I2 merck-NM_016223_at PACSIN3 merck2-NM_024103_x_at SLC25A23 merck-NM_005689_at ABCB6 merck-NM_182980_at OSGIN1 merck-ENST00000313594_x_at GCSH LOC101060817 merck-NM_006062_at SMYD5 merck2-NM_005035_at POLRMT merck-NM_001014832_s_at PAK4 merck2-BM970572_at OTUD7B merck-NM_001492_s_at CERS1 merck2-ENST00000358681_at EXT2 merck-NM_012476_at VAX2 ATP6V1B1 merck-NM_020378_at NAT14 merck2-AK026006_a_at TMEM53 merck-NM_004082_at DCTN1 merck2-NM_005789_at PSME3 AOC2 merck2-NM_014015_at merck2-AL832023_at POFUT1 merck-NM_017802_s_at HEATR2 merck-BC072383_s_at NPAS2 merck2-BC002515_s_at merck-CD014070_s_at TUBG2 merck-NM_001040716_at PC merck-NM_006690_s_at MMP24 merck2-CR600560_at EMC8 merck-NM_180976_at PPP2R5D merck-NM_015277_s_at NEDD4L merck-NM_178012_at TUBB2B merck2-AF059195_at MAFG merck-NM_001182_at ALDH7A1 PDE8B merck-NM_004422_at DVL2 ACADVL merck2-CK821133_a_at merck-NM_003780_at B4GALT2 merck-ENST00000334310_a_at TEAD1 merck-NM_005234_at NR2F6 merck2-AF147421_at ARHGAP5-AS1 merck-AY672105_a_at POLRMT CYP4F11 CYP4F2 merck-NM_016147_s_at PPME1 merck-NM_032829_at FAM222A merck-NM_152600_at ZNF579 merck-NM_001037131_at AGAP1 merck-NM_017797_s_at BTBD2 merck-BC005142_a_at AP3D1

Example 10 Prognostic Model for Soft Tissue Cancer

This example describes a soft tissue cancer prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model. Since both the prognosis signatures derived from the current dataset and the pre-defined proliferation signature predict patient outcome, both predictors were combined.

A total of 190 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model validated using the second half of samples. In the first half of samples, 261 samples had outcome data (live or death). In the first half of samples, 95 samples had outcome data (alive or dead). Among them, 49 had good outcome and 46 had poor outcome. 11 of the 49 good outcome patients did not have detailed last follow-up dates. In the second half of samples, all 95 had outcome data. Among them, 46 had good outcome and 49 had poor outcome. 5 out of the 46 good outcome patients did not have detailed follow-up dates.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 95 training samples which are either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 40 & 41.

A model was built in the training set using a general linear model (from the R package) using the following equation:


Soft Tissue Cancer Risk Score=Risk Score=0.39820+0.30357*(prg2−prg1)   (Formula 19),

where “prg1” is a score calculated from prognosis genes in Table 40 and “prg2” is a score calculated from prognosis genes in Table 41. The scores can be calculated by averaging the log2(intensity) of each probe in the geneset.

The performance of this model is evaluated in reserved validation set of 95 samples. FIG. 34 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 42.

TABLE 42 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.2 20 0 0.000 02.-0.4 29 14 0.483 0.4-0.6 20 13 0.650 >0.6 26 18 0.692

Using a threshold of 0.34, the odds ratio for overall survival is 6.9, 95% CI: 2.7-17.6, Fisher's Exact Test p-value=2.4×10−5.

Patients can be further divided into good (risk score<0.34), medium (score 0.34-0.55) and poor (score>0.55) prognosis groups. FIG. 35 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 18.3 (P=1.1×10′).

The number of genes in each pathway was reduced to 10 genes.

Prognosis Signature Component 1 (prg1):

    • Probe IDs: merck2-CN308012_at, merck-NM_003617_at, merck-NM_001981_at, merck-NM_014774_at, merck-NM_033439_at, merck-NM_017719_at, merck-NM_012158_at, merck2-AA551214_a_at, merck-BC030112_at, merck2-ENST00000377993_at

Gene symbols: EFCAB14, RGS5, EPS15, EFCAB14, IL33, SNRK, FBXL3, MBNL1, HIPK3, CMAHP

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck-CR407609_a_at, merck2-NM_005782_at, merck-BI084560_s_at, merck-BC066298_a_at, merck-ENST00000311926_s_at, merck-NM_003860_s_at, merck2-BM504304_a_at, merck2-XM_001134348_at, merck2-DC428989_at, merck-BG504479_s_at
    • Gene symbols: MRPS12, ALYREF, SNRPB, LSM12, UBE2S, BANF1, LSM4, ANAPC11, HNRNPK, RANBP1

The scores derived from these 10-genes are correlated to the original scores at the level of 0.92 for prg1, 0.94 for prg2.

Using the reduced gene sets, the updated predictive model is:


Soft Tissue Cancer Risk Score=0.74291+0.16726*(prg2−prg1)   (Formula 20).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

Patients in the validation set are grouped by the prediction score. Table 43 shows the detailed information about number of samples, number of deaths, and the death rate in each prediction score bin.

TABLE 43 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.2 12 2 0.167 0.2-0.4 26 9 0.346 0.4-0.6 32 22 0.688 >0.6 25 16 0.640

Using a threshold of 0.34, the odds ratio for overall survival is 7.4 (95% CI: 2.5-22.0), Fisher's Exact Test p-value=1.6×10−4.

Patients can be further divided into good (risk score<0.34), medium (score 0.34-0.55) and poor (score>0.55) prognosis groups. FIG. 36 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 16.1 (P=3.2×10-4).

A predefined proliferation signature (Table 44) is also prognostic in soft tissue cancer patients. The correlation of the proliferation score and the Risk Score of Formula 20 in soft tissue patients is 0.51.

The model was built in the training set using a general linear model (from the R package) with the following components:


Soft Tissue Cancer Risk Score=−0.32072+0.10405*pscore   (Formula 21).

Where pscore is the score calculated from prognosis genes in Table 44 by averaging the log2(intensity) of each probe in the geneset.

The performance of this model is evaluated in reserved validation set of 95 samples. FIG. 37 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 45.

TABLE 45 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.4 23 3 0.130 0.4-0.5 20 10 0.500 0.5-0.6 24 16 0.667 >0.6 28 20 0.714

Using a threshold of 0.42, the odds ratio for overall survival is 7.4, 95% CI: 2.5 -22.0, Fisher's Exact Test p-value=1.6×10−4.

Patients can be further divided into good (risk score<0.42), medium (score 0.42-0.55) and poor (score>0.55) prognosis groups. FIG. 38 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 16.8 (P=2.3×10−4).

The number of genes in proliferation signature can be reduced to 10 genes.

    • Probe IDs: merck-NM_012112_at, merck-NM_004701_at, merck-NM_001809_at, merck-NM_145060_at, merck-CR602926_s_at, merck-U63743_a_at, merck-NM_018101_at, merck2-AK000490_a_at, merck-NM_080668_at, merck-ENST00000333706_x_at
    • Gene symbols: TPX2, CCNB2, CENPA, SKA1, CCNB1, KIF2C, CDCA8, DEPDC1, CDCA5, BIRC5

The scores derived from these 10-genes are correlated to the original scores at the level of 0.99.

Using the reduced gene sets, the updated predictive model is:


Soft Tissue Cancer Risk Score=−0.24302+0.08483*pscore   (Formula 22).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

In the validation set, the detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 46.

TABLE 46 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.4 21 3 0.143 0.4-0.5 20 11 0.550 0.5-0.6 29 19 0.655 >0.6 25 16 0.640

Using a threshold of 0.40, the odds ratio for overall survival is 9.9 (95% CI: 2.7-36.5), Fisher's Exact Test p-value=1.3×10−4.

Patients can be further divided into good (risk score<0.4), medium (score 0.4-0.55) and poor (score>0.55) prognosis groups. FIG. 39 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 18.0 (P=1.2×10−4).

The two models (Formula 20 and Formula 22) can be combined to a single model to predict patient outcome. The combination can be done either by averaging the prediction scores, or by counting the risk factors.

FIG. 40 shows the Kaplan-Meier plot using the average risk score RS:


Soft Tissue Cancer Risk Score=(RS1+RS2)/2   (Formula 23).

Where RS1 is the risk score from Formula 20 and RS2 the risk score from Formula 22. When patients in the validation set were binned into three groups (<0.4, 0.4-0.55, and >0.55), the Chi-square on 2 degrees of freedom is 16.4 (P=2.7×10−4).

Alternatively, the risk scores from Formula 20 and Formula 22 can be first dichotomized into risk factors as:

RF1=1 if RS1>0.408, and RF1=0 if RS1<=0.408

RF2=1 if RS2>0.436, and RF2=0 if RS2<=0.436

RF=RF1+RF2

FIG. 41 shows the Kaplan-Meier plot for patients with RF ranges from 0 to 2. The Chi-square for 2 degrees of freedom is 19.6 (P=5.7×10−5).

TABLE 40 Prognosis signature component 1 (anti-correlated with poor outcome) probe Gene merck-NM_015208_at ANKRD12 merck-NM_005410_s_at SEPP1 CCDC152 merck-NM_013262_s_at MYLIP merck-NM_012096_at APPL1 merck-AK057337_at LINC00924 merck-AK091904_at merck-NM_000867_at HTR2B merck2-BX647414_a_at merck-NM_014774_at EFCAB14 merck-NM_003022_at SH3BGRL merck-BX647414_s_at merck2-CN371999_a_at FBXL3 merck2-AA155774_at RHOJ merck-AV703096_s_at merck-NM_031474_at NRIP2 merck-AK022074_a_at RUFY3 merck-NM_012158_at FBXL3 merck2-CN308012_at EFCAB14 merck2-NM_003922_at HERC1 merck-ENST00000375110_at EPC1 merck2-ENST00000367436_a_at CDC73 merck-BX647696_a_at TACC1 merck-BC036296_at merck-BF663662_at merck-AK022059_at SNX18 merck-AK092045_s_at CCDC50 merck-ENST00000368886_at IKZF5 merck-NM_194434_at VAPA merck2-CR623081_x_at merck2-AK223450_a_at MPPE1 GNAL merck-BX098521_at MAF LOC101928230 merck-NM_015602_a_at TOR1AIP1 merck2-DA809388_at CCDC50 merck2-NM_012158_at FBXL3 merck2-AF063564_x_at merck2-AF063564_at merck-AB008109_a_at RGS5 merck2-CD512895_at MYCBP2 merck2-AF030108_at RGS5 merck-ENST00000361850_at LINC00310 merck2-AI201749_x_at AR merck-NM_016089_at ZNF589 merck-NM_183419_s_at RNF19A merck-NM_003895_at SYNJ1 merck-NM_198159_at MITF merck2-AI201749_at AR merck-NM_033439_at IL33 merck-BC090936_at ZBTB20 merck2-BC013872_at TP73-AS1 merck-AF131806_at RGS3 merck-AW977864_at merck2-CA312624_at UQCRB merck2-N95413_at CREBL2 merck-NM_017831_at RNF125 merck-CR604678_s_at KRCC1 merck2-AL049423_at merck-AY007149_at CEP350 merck2-NM_024529_at CDC73 merck-AF147316_at merck-BC030112_at HIPK3 merck2-AL049787_at N4BP2L1 merck-NM_002022_at FMO4 merck-NM_005449_at FAIM3 IL24 merck2-NM_021140_at KDM6A CXorf36 merck-AL834204_a_at ANKRD12 merck2-CB852612_at SNX18 merck-NM_017719_at SNRK merck-NM_015346_at ZFYVE26 merck-BC039516_s_at merck2-NM_152267_at RNF185 merck2-NM_207292_at MBNL1 merck2-NM_031491_at RBP5 merck-NM_020940_s_at FAM160B1 merck2-BG701526_at merck-NM_000109_at DMD merck-BX648284_s_at ITGA1 merck2-NM_016302_at CRBN merck-NM_002697_a_at POU2F1 merck-CR595827_s_at PNRC2 merck-AK055652_at CCDC50 merck-NM_001025197_s_at CHI3L2 merck-NM_001289_at CLIC2 merck-AF086173_at TOR1AIP1 merck-NM_005149_at TBX19 merck-NM_001008390_at CGGBP1 merck-NM_032738_at FCRLA merck-AB011115_at ZNF862 merck-NM_015460_at MYRIP merck2-NM_032738_at FCRLA merck-BX648371_at LINC00861 merck-BM561378_at ACER3 merck2-DB317311_at GIMAP1 merck-NM_018105_at THAP1 merck2-AK129610_at SH3BGRL merck-AL832613_at SLC46A1 merck2-NM_023075_at MPPE1 GNAL merck2-AA551214_a_at MBNL1 merck-NM_024756_at MMRN2 merck-AK128852_a_at merck2-NM_080416_a_at

TABLE 41 Prognosis signature component 2 (correlated with poor outcome) probe Gene merck-BQ919512_s_at ALYREF merck-NM_198175_s_at NME1 merck2-NM_005782_at ALYREF merck-NM_001536_at PRMT1 merck2-AI654832_a_at ALYREF merck2-NM_033362_at MRPS12 merck2-DC428989_at HNRNPK merck-NM_172341_at PSENEN merck-NM_020438_at DOLPP1 merck2-BI602361_s_at merck2-BC002505_at SNRPF merck-CR407609_a_at MRPS12 merck-ENST00000311926_s_at UBE2S merck2-DA435913_at NCL merck-NM_003860_s_at BANF1 merck2-DA572591_a_at NCL merck-NM_005796_a_at NUTF2 CEP112 merck-NM_015179_s_at RRP12 merck-DA418198_s_at LARP1 merck-NM_052850_s_at GADD45GIP1 merck-NM_003707_s_at RUVBL1 merck-NM_001970_s_at EIF5AL1 EIF5A merck2-BX363921_x_at TOMM22 merck2-AL599091_x_at C5orf15 merck-NM_002809_at PSMD3 merck-NM_006428_at MRPL28 merck-NM_002949_at MRPL12 merck2-XM_001134348_at ANAPC11 merck-NM_003258_at TK1 merck-BI860175_a_at COQ4 merck-NM_032301_at FBXW9 merck2-BQ674733_at NUTF2 merck2-BM504304_a_at LSM4 merck-NM_016199_s_at LSM7 merck2-BM759128_a_at DDX54 merck-NM_144998_at STRA13 ASPSCR1 merck-BC025772_s_at EHMT1 merck-NM_002720_at PPP4C merck-NM_015679_at TRUB2 merck-ENST00000322030_x_at SET merck2-EF036485_at merck-NM_177542_at SNRPD2 merck-CR594938_s_at RRP1 merck2-AI809856_at RPL27A merck-BG771720_a_at EMC8 merck-NM_001002031_s_at ATP5G2 merck-CB995181_a_at LSM4 merck2-BG829700_at merck-NM_016034_at MRPS2 merck-NM_001833_at CLTA merck-NM_006114_s_at TOMM40 APOE merck-NM_032353_at VPS25 WNK4 merck2-CB122391_x_at merck-ENST00000306014_a_at DDX54 merck2-EF534308_x_at merck2-BG822880_x_at merck-CA866470_a_at RAD23B merck-NM_006808_at SEC61B merck-NM_017503_at SURF2 merck-BC066298_a_at LSM12 merck-CR596106_a_at CNPY2 merck-ENST00000355703_s_at PCNXL3 merck-ENST00000376263_a_at HNRNPK merck-AK057925_at CDKN2AIPNL merck2-NM_001040161_x _at C16orf13 merck2-CN304837_at PFDN2 merck-BC000118_at CLTA merck2-DB483456_at YWHAG merck2-CA848513_at CALR merck-AI911220_s_at VPS4A merck-NM_004870_at MPDU1 merck2-U28936_s_at merck-BC036909_at LOC284889 MIF merck-NM_025233_at COASY merck2-BC065000_a_at TCEB2 merck2-CD579847_at CALR merck2-AU132133_at UBE2Q2 merck-NM_006221_at PIN1 merck-AY735339_s_at CSNK2A1 CSNK2A3 merck-BM555073_s_at SNHG16 merck2-NM_003096_at SNRPG merck-ENST00000372692_s_at SET PARD3 merck-NM_006356_a_at ATP5H RAP1B merck2-CB122391_at merck2-BM755263_a_at YWHAE merck-NM_000990_x_at RPL27A merck2-BG748146_a_at FXN merck-NM_152383_s_at DIS3L2 merck-NM_006666_at RUVBL2 merck2-DA643319_at EHMT1 merck-NM_002904_a_at NELFE CFB merck2-NM_016050_a_at MRPL11 merck-NM_003310_at TSSC1 LOC101927554 merck-NM_006579_at EBP TBC1D25 merck-NM_014047_at C19orf53 merck2-BU623044_at ERCC2 merck-NM_175614_at NDUFA11 merck-BP224564_a_at YY1 merck-XM_939690_at RPS15P9 merck2-AA081397_x_at

TABLE 44 Proliferation signature probe Gene merck-NM_003318_at TTK merck-NM_014791_at MELK merck-NM_001786_a_at CDK1 RHOBTB1 merck-NM_001790_at CDC25C merck-NM_014176_at UBE2T merck-BF511624_s_at BUB1B merck-NM_005030_at PLK1 merck-NM_181802_at UBE2C merck-NM_004217_at AURKB merck-NM_201567_at CDC25A merck-NM_198436_s_at AURKA merck-NM_001255_s_at CDC20 merck-NM_003579_at RAD54L merck-NM_004336_at BUB1 RGPD6 merck-NM_031299_at CDCA3 GNB3 merck-NM_004237_at TRIP13 merck-BC001459_s_at RAD51 merck-NM_012484_at HMMR merck-AB042719_a_at MCM10 merck-NM_018518_at MCM10 merck-NM_012291_at ESPL1 PFDN5 merck-NM_014750_at DLGAP5 merck-NM_199413_at PRC1 merck-NM_130398_at EXO1 merck-NM_199420_s_at POLQ merck-NM_005733_at KIF20A CDC23 merck-NM_004856_at KIF23 merck-NM_004701_at CCNB2 merck-NM_014321_at ORC6 merck-NM_002466_at MYBL2 merck-NM_030919_at FAM83D merck-NM_003504_at CDC45 merck-BC075828_a_at GTSE1 merck-NM_016426_at GTSE1 TRMU merck-NM_001012409_at SGOL1 merck-NM_018136_s_at ASPM merck-NM_018685_at ANLN merck-NM_012112_at TPX2 merck-NM_018101_at CDCA8 merck-NM_001237_a_at CCNA2 EXOSC9 merck-NM_018454_at NUSAP1 merck-NM_001211_at BUB1B merck-U63743_a_at KIF2C merck-CRS96700_a_at RRM2 merck-NM_012310_at KIF4A GDPD2 merck-NM_013277_a_at RACGAP1 merck-NM_018154_at ASF1B PRKACA merck-BC024211_a_at NCAPH merck-NM_152515_at CKAP2L merck-NM_018131_at CEP55 merck-NM_002417_at MKI67 merck-CR607300_a_at MKI67 merck-BI868409_a_at MKI67 merck-NM_001813_at CENPE merck-CR602926_s_at CCNB1 merck-NM_001809_at CENPA merck-NM_080668_at CDCA5 merck-AK223428_a_at BIRC5 merck-NM_005480_at TROAP merck-NM_021953_at FOXM1 merck-NM_144508_at CASC5 merck-NM_019013_at FAM64A PITPNM3 merck-hCT1776373.2_s_at DEPDC1 OTUD7A merck-NM_004091_at E2F2 merck-NM_004219_x_at PTTG1 merck-NM_002263_a_at KIFC1 merck-AF331796_a_at NCAPG merck-NM_145060_at SKA1 merck-BC048988_a_at SKA3 merck-NM_152259_s_at TICRR KIF7 merck-ENST00000243201_a_at HJURP merck-ENST00000333706_x_at BIRC5 merck-ENST00000335534_s_at KIF18B merck-AY605064_at CLSPN merck2-AK097710_at CDC25C merck2-AF043294_at BUB1 RGPD6 merck2-AU132185_at MKI67 merck2-BC098582_at KIF14 merck2-BT006759_at KIF2C merck2-BC006325_at GTSE1 TRMU merck2-BC006325_x_at GTSE1 TRMU merck2-AL832036_at CKAP2L merck2-DQ890621_at CDC45 merck2-NM_005196_at CENPF merck2-AV714642_at ANLN merck2-BC034607_at ASPM merck2-BC001651_at CDCA8 merck2-AF098158_at TPX2 merck2-NM_001168_at BIRC5 merck2-AK023483_at NUSAP1 merck2-NM_145061_at SKA3 merck2-NM_018410_at HJURP merck2-AL517462_s_at merck2-ENST00000333706_s_at merck2-BX648516_at SGOL1 merck2-AK000490_a_at DEPDC1 merck2-ENST00000370966_a_at DEPDC1 OTUD7A merck2-AB046790_at CASC5 merck2-CR936650_at ANLN merck2-AL519719_a_at BIRC5 merck2-NM_145060_a_at SKA1 merck2-NM_001039535_a_at SKA1

Example 11 Prognostic Model for Uterus

This example describes a uterus prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

A total of 342 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model validated using the second half of samples. In the first half of samples, 168 samples had outcome data (alive or dead). Among them, 119 had good outcome and 49 had poor outcome. One good outcome patient did not have stage data. In the second half of samples, all 171 had outcome data. Among 130 good outcome patients, 13 did not have stage data. In the 41 poor outcome patients, 5 did not have stage data.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 168 training samples which are either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 47 & 48.

A model was built in the training set using a general linear model (from the R package) using the following equation:


Uterus Cancer Risk Score=0.33692+0.10294*(prg2−prg1)+0.09746*stage   (Formula 24),

where “prg1” is a score calculated from prognosis genes in Table 47 and “prg2” is a score calculated from prognosis genes in Table 48. The scores can be calculated by averaging the log2(intensity) of each probe in the geneset.

The performance of this model is evaluated in reserved validation set of 153 samples with also the stage data. FIG. 42 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 49.

TABLE 49 Average death rate versus prediction score. Score Number of samples Number of death Death Rate <0.2 61 5 0.082 0.2-0.4 46 7 0.152 0.4-0.6 32 15 0.469 >0.6 14 9 0.643

Using a threshold of 0.4, the odds ratio for overall survival is 9.3, 95% CI: 3.8 -22.5, Fisher's Exact Test p-value=1.1×10−7.

Patients can be further divided into good (risk score<0.32), medium (score 0.32-0.6) and poor (score>0.6) prognosis groups. FIG. 43 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 40 (P=2.1×10−9).

The number of genes in each pathway was reduced to 10 genes.

Prognosis signature component 1 (prg1):

    • Probe IDs: merck-ENST00000369936_at, merck-NM_004058_at, merck-NM_002407_at, merck-AI918006_at, merck2-AK025905_at, merck-NM_145051_s_at, merck2-DT217746_at, merck-NM_152376_s_at, merck-NM_006551_at, merck2-CA489714 at
    • Gene symbols: KIAA1324, CAPS, SCGB2A1, UBX1V10, SOX17, RNF183, ASRGL1, UBXN10, SCGB1D2, SPDEF

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck2-BM904739_at, merck-NM_153485_at, merck-NM_003875_at, merck-NM_000540_at, merck-NM_021922_at, merck-NM_181573_s_at, merck-ENST 00000311926_s_at, merck2-BC112898_at, merck-NM_007274_s_at, merck-NM_004181_at
    • Gene symbols: MRGBP, NUP155, GMPS, RYR1, FANCE, RFC4, UBE2S, ZNF623, ACOT7, UCHL1

The scores derived from these 10-genes are correlated to the original scores at the level of 0.97 for prg1, 0.94 for prg2.

Using the reduced gene sets, the updated predictive model is:


Uterus Cancer Risk Score=0.15030+0.06071*(prg2−prg1)+0.10849*stage (Formula 25).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

FIG. 44 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate for this updated model. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 50.

TABLE 50 Average death rate versus prediction score. Number os Number of Score samples death Death Rate <0.2 63 6 0.095 0.2-0.4 44 7 0.159 0.4-0.6 34 14 0.412 >0.6 12 9 0.750

Using a threshold of 0.32, the odds ratio for overall survival is 8.5 (95% CI: 3.5-20.6), Fisher's Exact Test p-value=4.1×10−7.

Patients can be further divided into good (risk score<0.32), medium (score 0.32-0.6) and poor (score>0.6) prognosis groups. FIG. 45 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 40.9 (P=1.3×10−9).

TABLE 47 Prognosis signature component 1 (anti- correlated with poor outcome) Probe Gene merck-AL040975_at ESR1 merck-NM_005397_at PODXL MKLN1 merck-AI918006_at UBXN10 merck-AL137566_at PGR merck-NM_022454_at SOX17 merck2-AA148029_at PODXL MKLN1 merck2-AK025905_at SOX17 merck-NM_002407_at SCGB2A1 merck-NM_001012993_at C9orf152 merck2-NM_000125_at ESR1 merck-NM_000125_at ESR1 merck-NM_018728_at MYO5C merck2-AL050116_at ESR1 merck-AF016381_a_at PGR merck-BX106921_at PGR merck-NM_006551_at SCGB1D2 merck-BX648070_at C2orf88 HIBCH merck-ENST00000369936_at KIAA1324 merck-NM_152376_s_at UBXN10 merck-NM_014178_s_at STXBP6 merck2-BX648631_at UBXN10 merck-BC028018_at LOC100129098 merck2-BQ684833_at ACSL5 merck-NM_014211_at GABRP merck-NM_021069_at SORBS2 merck-BC011052_a_at TRIM2 merck-AL834346_at STXBP6 merck-ENST00000347491_s_at ESR1 merck2-DT217746_at ASRGL1 merck-NM_004058_at CAPS merck-NM_025080_s_at ASRGL1 merck-NM_005080_at XBP1 merck-NM_018414_at ST6GALNAC1 merck-NM_020775_s_at KIAA1324 merck2-AM392558_at SORBS2 merck-ENST00000319471_a_at SORBS2 merck2-NM_021777_at ADAM28 merck-NM_015541_s_at LRIG1 merck-ENST00000285039_at MYO5B merck-NM_002644_s_at PIGR merck2-CB852618_at GRAMD3 merck2-NM_016930_at STX18 merck-BC017958_at CCDC160 merck-NM_013992_at PAX8 merck-NM_174921_at SMIM14 merck-NM_003212_at TDGF1 merck2-CA489714_at SPDEF merck2-BG742453_a_at PAM merck-AJ420553_at ID4 merck-NM_138766_s_at PAM merck2-AF137334_at ADAM28 merck-NM_001669_at ARSD merck2-NM_014133_at SORBS2 merck-NM_175887_at PRR15 merck-NM_018050_at MANSC1 merck2-CB241906_at ST6GALNAC1 merck-ENST00000369949_s_at C1orf194 merck-AL702564_at PGR merck-NM_001025593_at ARFIP1 merck-NM_018043_at ANO1 merck-NM_012391_at SPDEF merck-NM_021785_at RAI2 merck-NM_014265_at ADAM28 merck2-BC008590_at GRAMD3 merck2-CB962832_at ID4 merck-NM_003774_at POC1B-GALNT4 GALNT4 merck-NM_015271_at TRIM2 merck-AK128437_a_at GALNT7 merck2-BM695584_at ARHGAP26 merck-NM_001004303_at C1orf168 merck-BC094795_a_at PIK3R1 merck-NM_015071_at ARHGAP26 merck-NM_145051_s_at RNF183 merck-NM_001915_at CYB561 merck-AW970730_at ST6GALNAC1 merck-BC002976_s_at CYB561 merck-NM_015198_at COBL merck-CA427248_at CCDC122 merck-NM_001490_at GCNT1 merck-NM_022783_at DEPTOR merck2-AK026697_at CDS1 merck-NM_020879_s_at CCDC146 merck-NM_001040001_at MLLT4 KIF25 merck-NM_032321_a_at C2orf88 merck2-NM_033087_at ALG2 merck-NM_001006615_s_at WDR31 merck-NM_030630_s_at HID1 merck-NM_153000_at APCDD1 merck-NM_176813_at AGR3 merck-CR749204_s_at PTPN3 merck-NM_000266_at NDP merck-NM_004727_s_at SLC24A1 merck2-BC012630_at SLC24A1 merck-NM_015993_at PLLP merck-BC068555_a_at ARHGAP26 merck-T68445_a_at AR merck-NM_001002912_s_at C1orf173 merck2-AK023916_at DEPTOR merck-AB032983_at PPM1H merck-AK075059_at GLIS3

TABLE 48 Prognosis signature component 2 (correlated with poor outcome) Probe Gene merck2-AB071393_a_at TTL merck2-AK127448_at B4GALNT1 merck2-NM_153712_at TTL merck-NM_001010911_at CASC10 merck2-BM904739_at MRGBP merck-NM_000540_at RYR1 merck-NM_006442_s_at DRAP1 merck2-AK222554_x_at SF3A3 merck-BU594972_a_at TSC1 merck-CR599730_a_at TTL merck2-BU620949_at DRAP1 merck2-AK222554_at SF3A3 merck-BC029828_at B4GALNT1 merck-NM_003875_at GMPS merck-ENST00000222607_at STEAP1B merck-NM_006143_at GPR19 merck2-BC112898_at ZNF623 merck-NM_021922_at FANCE merck2-BI602361_s_at merck-AL832168_at merck2-AI825916_at TSC1 merck2-BC041955_at merck2-NM_199427_at ZFP64 merck2-AI149996_at ADRM1 merck-NM_004181_at UCHL1 merck-NM_181573_s_at RFC4 merck-BC028609_a_at CCDC93 merck-AF368281_a_at SGTB merck-ENST00000311926_s_at UBE2S merck-NM_021158_at TRIB3 merck-NM_006087_at TUBB4A merck2-AK026140_at merck2-AK130014_at SHC1 merck-NM_003610_at RAE1 merck-NM_018270_at MRGBP merck-NM_016447_at MPP6 merck-NM_182627_at WDR53 merck-AL713706_at DPYSL5 merck-NM_014696_s_at GPRIN2 merck-AB015342_a_at ZNF318 merck2-ENST00000356433_at DLL3 merck2-BF739910_at RBM33 merck-NM_004341_at CAD merck-ENST00000313019_s_at SHOX2 merck-BC003580_s_at CIAO1 merck-NM_001426_at EN1 merck-NM_002503_at NFKBIB merck-NM_016625_s_at RSRC1 merck2-DA447204_at SHOX2 merck-AF533230_x_at USP32 merck-NM_013409_at FST merck2-BC012379_at ZHX1-C8ORF76 merck-NM_007274_s_at ACOT7 merck-AK123535_at FBXL18 merck-NM_152699_s_at SENP5 merck-NM_007002_at ADRM1 merck2-BC025263_at CDCA4 merck-NM_006553_at SLMO1 merck-NM_206831_a_at DPH3 OXNAD1 RFTN1 merck-NM_006818_at MLLT11 merck-NM_000523_at HOXD13 merck-AK025697_at FBXO45 merck2-BX340398_at SMIM13 merck-AW821325_at RAE1 merck2-BC001395_at CIAO1 merck-BT009760_s_at ZFP64 merck-NM_000022_at ADA merck-DW451489_s_at MED8 merck2-NM_001017406_at S100PBP merck-ENST00000343379_a_at SS18L1 merck2-BC051770_a_at ACTN2 merck-AK129880_a_at UBXN7 merck-BC064390_a_at HAUS5 merck-NM_001039617_at ZDHHC19 merck2-NM_145733_at 3-Sep merck-BC068057_a_at YRDC merck2-NM_023008_at KRI1 merck2-BC040609_at SENP2 merck2-AB053301_at TMEM237 merck-NM_007027_at TOPBP1 merck-NM_001008949_at ITPRIPL1 merck-NM_178830_at C19orf47 merck-NM_183001_a_at SHC1 merck-AF151697_a_at SENP2 merck-ENST00000362037_at LOC645195 merck-NM_012318_at LETM1 merck-NM_153485_at NUP155 merck-NM_002808_at PSMD2 merck-BC047330_at MPP6 merck-NM_024333_at FSD1 STAP2 merck-NM_152363_at ANKLE1 merck-AK126101_a_at PLXNA1 merck2-AB209521_at ACTN2 merck-NM_015327_at SMG5 PTS merck2-BM674474_at merck-BC014211_x_at TCEA2 merck-NM_024721_a_at ZFHX4 merck-BC042486_a_at KIF3C merck-NM_203486_s_at DLL3 merck-NM_001350_s_at DAXX

Example 12 Prognostic Model for Ovarian Cancer

This example describes an ovarian cancer prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model. Since both the prognosis signatures derived from the current dataset and the pre-defined proliferation signature predict patient outcome, both predictors were combined.

A total of 731 samples were profiled by Affymetrix® expression arrays. Among them 362 were alive and 367 were dead (2 with status unknown) at the time of data collection. Samples were equally divided into training (365 samples) and validation (366 samples) set. In the training set, patients were first divided into two groups based on genome-wide 2-D clustering, and the markers associated with these two groups were identified. Among the markers correlated with group IDs, one group of markers (X2) led to successful prognosis biomarker identification when used in the patient stratification.

In the training set, a 2D-clustering based on 3171 highly variable genes (standard deviation of log2 intensity)>1.5) was performed, and patients were partitioned into two groups. Genes were then selected that are highly variable (std(log2 intensity)>2) and with correlation to the group ID greater than 0.5 (positive- and negative-correlation). Each group of genes was used to stratify patients for prognosis, and a group of genes (listed in Table 51) enabled discovery of strong prognosis patterns in the training set.

TABLE 51 patient stratification markers Correlation to Probe ID Gene group ID merck-AI732822_at KCND2 0.523155 merck2-AI264554_at 0.543379 merck-BX103595_at 0.580491 merck-NM_015507_at EGFL6 0.541111 merck-NM_001878_at CRABP2 0.526755 merck-NM_012427_at KLK5 0.54748 merck-NM_005046_s_at KLK7 0.554217 merck-NM_016725_s_at FOLR1 0.502639 merck-NM_001276_at CHI3L1 0.506725 merck-ENST00000373692_a_at PTGS1 0.582718

Patient stratification was based on the average log2 intensity from the probes listed in Table 51. FIG. 46 shows the histogram of the X2 probe intensities in ovarian cancer. There is peak around log2 intensity of 10, and a uniform distribution below the intensity peak. When the X2 intensity versus the estrogen-receptor level was checked, almost all the patients with high X2 intensity also had uniformly high ER intensity, contrasting to the low-X2 patients where ER levels had wide range (FIG. 47). A threshold was therefore placed at X2 =9. Patients with X2>9 and X2<9 will be termed X2+ and X2− in the rest of the example.

In the training set with 365 samples, 175 patients had X2− (X2<9), and 190 patients with X2+ (X2>9). In the X2−, 174 patients had outcome data, 88 were dead at the time of data collection. In the X2+ patients, 189 had outcome data, 118 were dead. Prognosis signature discovery was tried for both X2− and X2+ populations. For this example, the focus is on X2− since it yielded a more significant prognostic model.

In the validation set with 366 samples, 170 patients are X2− and 196 patients are X2+. The poor outcome patients (dead at the last time of data collection) are 75 and 86 respectively.

Patients with high X2 had slightly higher poor outcome rate, but X2 itself is not a strong prognosis factor.

Two groups of genes (100 Affymetrix® probe-sets each) were identified in 174 X2− raining samples which are either correlated or anti-correlated with poor outcome. These two groups of genes are displayed in Tables 52 & 53.

A model was built in the X2− training set using a general linear model (from the R package) using the following equation:


Ovarian Cancer Risk Score=−0.01678−(0.09271*prg1)+(0.10882*prg2)+(0.17827*stage)   (Formula 26),

where “prg1” is a score calculated from prognosis genes in Table 52 and “prg2” is a score calculated from prognosis genes in Table 53, and the stage is the composite stage. The scores can be calculated by averaging the log2(intensity) of each probe in the geneset.

The performance of this model is evaluated in reserved validation set of 170 X2− samples. FIG. 48 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate. As shown in the Figure, the model predicts the average death rate very well.

The detailed information about number of samples, number of deaths, and the death rate in each prediction score bin are summarized in Table 54.

TABLE 54 Average death rate versus prediction score. Number of Number of Score samples death Death Rate <0.2 23 0 0.000 0.2-0.4 25 4 0.160 0.4-0.6 27 11 0.407  0.6-0.08 50 30 0.600 >0.8 35 27 0.771

Using a threshold of 0.5, the odds ratio for overall survival is 9.6 (95% CI: 4.1-22.4), Fisher's Exact Test p-value=6.2×10−9.

Patients can be further divided into good (risk score<0.5), medium (score 0.5-0.7) and poor (score>0.7) prognosis groups. FIG. 49 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 34.3 (P=3.6×10−8).

In the prognosis model, two components are based signatures, and one component based on tumor stage. The signatures and tumor stage had similar prognosis power in the validation set. FIGS. 50A and 50B shows the prediction based on the signature only (using Formula 26 but drop the stage component) and tumor stage only. The predictive powers are very similar (Chi-squares on 2 degree of freedom are 34 for the signatures and 27.9 for the tumor stage).

The number of genes in each signature can be reduced to 10 genes.

Prognosis Signature Component 1 (prg1):

    • Probe IDs: merck-NM_025145_at, merck-AB051484_at, merck-NM_018430_s_at, merck-NM_018897_at, merck-NM_145170_at, merck-NM_181643_at, merck-NM_031421_at, merck-NM_003551_at, merck-NM_024763_at, merck-NM_178452_s_at
    • Gene symbols: WDR96, DNAH6, TSNAXIP1, DNAH7, TTC18, PIFO, TTC25, NME5, WDR78, DNAAF1

Prognosis Signature Component 2 (prg2):

    • Probe IDs: merck-NM_021972_at, merck2-BQ002341_at, merck2-NM_007115_at, merck-NM_004460_at, merck-NM_000960_at, merck-NM_002658_at, merck-X77690_at, merck-BC007858 a_at, merck-NM_003485_at, merck-AY358331sat
    • Gene symbols: SPHK1, LINC00607, TNFAIP6, FAP, PTGIR, PLAU, TIMP3, INHBA, GPR68, NTM

The scores derived from these 10-genes are correlated to the original scores at the level of 0.96 for prg1, 0.91 for prg2.

Using the reduced gene sets, the updated predictive model is:


Ovarian Cancer Risk Score=0.26269−(0.06569*prg1)+(0.03415*prg2)+(0.18904*stage)   (Formula 27).

Note, the exact coefficients will change depending on the final selection of the technology platform (RNAseq vs. arrays, PCR), and the probe sets or gene lists.

FIG. 51 shows the predicted death rate vs. the actual average (running average of 50 samples as ranked by the prediction score) death rate for this updated model. As shown in the Figure, the model predicts the average death rate very well.

Table 55 shows the detailed information about number of samples, number of deaths, and the death rate in each prediction score bin.

TABLE 55 Average death rate versus prediction score. Number of Number of Score samples death Death Rate <0.2 22 0 0.000 0.2-0.4 23 3 0.130 0.4-0.6 33 12 0.364  0.6-0.08 46 31 0.674 >0.8 36 26 0.722

Using a threshold of 0.5, the odds ratio for overall survival is 9.2 (95% CI: 4.1-20.9), Fisher's Exact Test p-value=4.0×10−9.

Patients can be further divided into good (risk score<0.5), medium (score 0.5-0.7) and poor (score>0.7) prognosis groups. FIG. 52 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 30.7 (P=2.1×10−7).

X2− and X2+ patients have different immune signature scores (FIGS. 53A and 53B), X2− patients have more spread but majority had low scores, whereas X2+ is peaked higher. When checking the outcome with immune scores, there is no relation between patient outcome and immune signature score in X2-patients, but in X2+ patients, high immune score is related to relative good outcome (P-value=1.2%).

X2 is highly correlated with keratins, and cadherins, and to a certain degree, with integrins as well (FIG. 54). For example, the correlation between X2 and the average of all keratins is 0.59. Clustering based all cadherins almost perfectly segregates X2+ from X2− patients. Among the cadherins, CDH6 is correlated to X2 at 0.61. Hence, X2+ may indicate tumors were originated from more “epithelial-like” tissues.

Table 56 lists the histotype distribution between X2− ad X2+ populations. X2− is enriched for Carcinosarcoma, Clear cell adenocarcinoma, Endometroid adenocarcinoma, Granulosa cell tumor and Mucinous adenocarcinoma, whereas X2+ is enriched for Papillary serous cystadenocarcinoma and Serous cystadenocarcinoma.

TABLE 56 Number of samples in X2− and X2+ population X2− X2+ Adenocarcinoma, NOS 29 31 Carcinoma, NOS 15 27 Carcinosarcoma, NOS 8 0 Clear cell adenocarcinoma, NOS 21 0 Endometrioid adenocarcinoma, NOS 35 7 Granulosa cell tumor, malignant 32 0 Mucinous adenocarcinoma 10 0 Papillary serous cystadenocarcinoma 46 106 Serous cystadenocarcinoma, NOS 76 206 Serous, borderline 12 0

When the disclosed endometrium cancer prognosis signature is applied to the ovarian cancer, the performance is significantly different in X2− and X2+ populations (FIG. 55A and 55B). In X2-population, the endometrium signature is a very strong predictor (chi-square=82.5, P=0), but same model is only marginally predictive in X2+ population (chi-square=4.3, P=0.04), suggesting X2− is more “endometrium-like”.

TABLE 52 Prognosis signature component 1 (anti- correlated with poor outcome) Probe Gene merck-NM_003551_at NME5 merck2-BC026182_at NME5 merck-NM_130897_at DYNLRB2 LOC101928276 merck-NM_003462_at DNALI1 merck-AF006386_a_at DNALI1 merck-AK055990_at DNAH9 merck-NM_145170_at TTC18 merck2-AB014543_at CLUAP1 merck2-BX093691_at TTC18 merck-ENST00000369736_a_at PIFO merck2-AI167680_a_at CLUAP1 merck-NM_018430_s_at TSNAXIP1 merck-NM_015041_a_at CLUAP1 merck-NM_152676_at FBXO15 merck-NM_181643_at PIFO merck2-XM_294004_at RSPH4A merck2-NM_001039845_at MDH1B merck-NM_031294_s_at LRRC48 ATPAF2 merck-NM_053000_s_at EPB41L4A-AS1 merck-NM_022785_s_at EFCAB6 merck-NM_145047_s_at OSCP1 merck-NM_024549_s_at TCTN1 merck-NM_014433_at RTDR1 merck2-BC034669_at DPH5 merck-AB051484_at DNAH6 merck-ENST00000341790_a_at NME9 merck-ENST00000374412_a_at MDH1B merck-G36659_at FANK1 merck-NM_001010892_at RSPH4A merck-NM_007081_s_at RABL2A RABL2B merck-NM_015958_s_at DPH5 merck2-AF546872_at PACRG merck-BC017958_at CCDC160 merck-NM_024763_at WDR78 merck2-NM_006961_at ZNF19 merck-AK027161_at TTC12 merck-NM_013249_at ZNF214 merck-NM_001551_at IGBP1 merck-NM_145235_at FANK1 merck-NM_152410_at PACRG merck2-NM_001100873_at C16orf46 CMC2 merck-NM_025145_at WDR96 merck-NM_176677_at NHLRC4 merck2-BC062574_at NPHP1 merck-NM_001008226_at FAM154B merck-U79257_at merck-NM_032257_s_at ZMYND12 merck2-BQ576016_at ZNF214 merck-CR593886_a_at RABL5 merck2-BC043273_at HYDIN merck-BU681848_a_at FLJ37035 LOC283038 merck2-AY336746_at NME9 merck2-AK093204_at DALRD3 WDR6 merck-BX648527_at TMEM232 merck-BE044185_a_at KIF6 merck2-BU785445_at ZMYND12 merck2-NM_206837_at OSCP1 merck-BC040979_at LINC00271 merck-BX647542_s_at PHKA1 merck2-BM977387_at merck2-CA426602_s_at merck-NM_001031745_at RIBC1 HSD17B10 merck-ENST00000303697_at DCDC5 merck-BX571745_a_at NPHP1 merck-NM_152572_at AK8 merck2-BC029902_at LRRC27 merck-NM_022784_at IQCH merck-AL832607_s_at SPEF2 merck2-NM_000967_s_at merck2-CA426602_at LRRC6 merck2-BC047091_a_at ZNF19 merck-BC058159_a_at LRRC27 merck-NM_024608_at NEIL1 MAN2C1 merck-NM_207417_at C9orf171 merck-NM_017775_at TTC19 merck-NM_175885_at FAM181B merck-NM_178832_s_at MORN4 merck2-AA481616_at merck2-AK125886_at merck-BC017993_at SNHG8 merck2-DR159121_at FBXO21 merck-NM_022777_at RABL5 merck-NM_015002_at FBXO21 merck-ENST00000341761_at WDR31 merck-NM_080667_s_at CCDC104 merck2-AL833327_at DNAAF1 merck2-AW959853_at ATXN10 merck-NM_018897_at DNAH7 merck-AL137566_at PGR merck-NM_001006615_s_at WDR31 merck2-BC007345_at RPL13 merck2-BC007345_x_at RPL13 merck-NM_004650_at PNPLA4 merck-NM_024867_s_at SPEF2 merck-NM_012119_at CDK20 merck2-AA383024_s_at merck-NM_194270_at MORN2 merck2-BC031231_at STK33 merck2-BC033935_at FBXO36 merck-AK097547_s_at SPEF2

TABLE 53 Prognosis signature component 2 (correlated with poor outcome) probe Gene merck2-AK127448_at B4GALNT1 merck-NM_021972_at SPHK1 merck-NM_003942_at RPS6KA4 merck-BC007582_a_at CEBPG merck-NM_000960_at PTGIR merck2-BQ002341_at LINC00607 merck2-NM_004145_at MYO9B merck2-BX340398_at SMIM13 merck-ENST00000332498_x_at CYCSP3 merck-NM_022338_at C11orf24 merck-X77690_at TIMP3 merck-BC005339_a_at TPMT merck-NM_004521_s_at KIF5B merck2-AK027899_a_at RELT merck2-NM_003039_at SLC2A5 merck-BC051810_a_at RELT merck-NM_138441_s_at MB21D1 merck2-D45917_a_at TIMP3 merck2-NM_007115_at TNFAIP6 merck-NM_024656_at COLGALT1 merck2-AI537528_x_at TUBA1B merck-BC071897_a_at MCL1 merck-AF006082_a_at ACTR2 merck2-AB030656_at CORO1C merck-DW451489_s_at MED8 merck-AW072050_a_at MYO9B merck-AY177688_s_at DNAJC21 merck-NM_002524_at NRAS merck-NM_054034_a_at FN1 merck-NM_002928_at RGS16 merck-NM_006884_s_at SHOX2 merck-M31164_at TNFAIP6 merck-AF143684_s_at MYO9B merck2-AF456425_a_at DCUN1D1 merck-NM_005192_at CDKN3 merck2-CA308717_at merck-CR627287_at ALDH1L2 merck-BC073853_a_at ACER3 merck-AY171233_s_at PTPDC1 merck2-AX801509_a_at TIMP3 merck-AI160141_a_at SLC2A5 merck-NM_030759_a_at NRBF2 merck-NM_002202_at ISL1 merck2-AA661461_at TUBA1B merck2-AI566394_at COLGALT1 merck2-AA758689_at SKIL merck-NM_015459_s_at ATL3 merck2-ENST00000378047_at FGF1 merck-CR610281_a_at TIMP3 merck-NM_001189_at NKX3-2 merck-ENST00000284274_a_at FAM105B merck-81258956_a_at PTBP3 merck2-AK097588_at ATL3 merck-NM_021958_at HLX merck2-BX096261_a_at SLC2A5 merck-NM_016573_at GMIP merck-BC029828_at B4GALNT1 merck-NM_004226_at STK17B merck2-BC032912_at NADK2 merck-NM_006101_at NDC80 merck2-BM740515_at merck-NM_014632_s_at MICAL2 merck-NM_002093_at GSK3B merck-NM_015719_at COL5A3 merck-NM_001945_at HBEGF merck2-BI824983_a_at ACER3 merck-NM_004994_at MMP9 merck-BC032697_a_at FGF1 merck2-NM_001031800_at TIPRL merck2-NM_004994_at MMP9 merck-CD106390_s_at RAP1A merck-BC006243_a_at RGS16 merck2-CR594502_at TIMP3 merck-BC035724_a_at NAB1 merck-NM_005261_at GEM merck-NM_001034173_a_at ALDH1L2 merck-NM_025217_at ULBP2 merck-NM_145805_at ISL2 merck-AJ419936_a_at TNFAIP6 merck-CR619305_a_at GNB1 merck-NM_024947_at PHC3 merck-NM_178167_a_at ZNF598 merck-NM_004460_at FAP merck2-BC028284_at MARCKS HDAC2 merck-CB529742_at merck-NM_001009936_a_at PHF19 merck-BC087859_at LOC401317 merck-NM_018304_s_at PRR11 merck-AU121101_a_at THBS2 LOC1011929523 merck-NM_005990_at STK10 merck-G36532_at TIMP3 merck-XM 292021_at SMCO2 merck-NM_032505_at KBTBD8 merck-NM_016287_at HP1BP3 merck-NM_005651_at TDO2 merck2-AI732388_at MGAT4A merck2-BC126107_a_at TEP1 merck2-BX349325_at PRR11 merck-NM_001747_at CAPG AFFX-HSAC07/X00351_3_at ACTB

Example 13 Prognostic Model for Bladder Cancer

This example describes a bladder cancer prognosis model based on gene expression profiling data. The model contains two gene expression signatures as components. In the second part of the example, the number of genes in each signature is reduced to 10 genes to simplify the implementation of this prognosis model.

A total of 273 samples were profiled by Affymetrix® expression arrays. A composite model was built using the first half of samples and the model validated using the second half of samples. In the training set, 137 samples had outcome data (alive or death). In the validation set, 136 had outcome data. The detailed last follow-up dates for the good outcome patients are incomplete. In the training set, 18 out of 47 good outcome patients did not have the last follow-up date. In the validation set, 4 out of 37 good outcome patients did not have the last follow-up date.

A model was built in the training set using a general linear model (from the R package) using the following equation:


Bladder Cancer Risk Score=0.60864−(0.06571*imscore)+(0.06168*hscore)   (Formula 27),

where imscore is the immune signature score calculated from signature genes in Table 57 and hscore is the hypoxia signature score calculated from signature genes in Table 58. The scores can be calculated by averaging the log2(intensity) of each probe in the geneset.

The performance of this model is evaluated in reserved validation set of 136 samples. Table 59 lists number of samples, number of deaths, and the death rate in each prediction score bin.

TABLE 59 Average death rate versus prediction score. Number of Number of Score samples death Death Rate <0.6 22 11 0.50 0.6-0.7 38 26 0.68 0.7-0.8 46 37 0.80 >0.8 30 25 0.83

Using a threshold of 0.66, the odds ratio for overall survival is 4.4 (95% CI: 2.0-9.8), Fisher's Exact Test p-value=3.4×10−4.

Patients can be further divided into good (risk score<0.66), medium (score 0.66-0.75) and poor (score>0.75) prognosis groups. FIG. 56 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 13.3 (P=1.3×10−3).

The number of genes in each pathway can be reduced to 10 genes.

Immune signature:

    • Probe IDs: merck-NM_002209_at, merck2-BI519527_at, merck-NM_000733_at, merck-NM_001778_at, merck2-NM_052931_at, merck-NM_001767_at, merck-NM_198517_at, merck-NM_024070_at, merck-NM_014207_at, merck-NM_032214_at
    • Gene symbols: ITGAL, IKZF1, CD3E, CD48, SLAMF6, CD2, TBC1D10C, PVRIG, CD5, SLA2

Hypoxia Signature:

    • Probe IDs: merck2-NM_005555_at, merck2-X56807_at, merck-BX538327_at, merck-XM_928117_x at, merck2-NM_005554_at, merck-AL572710_s_at, merck-NM_006945_at, merck-X15014_a_at, merck2-AI989728_at, merck-NM_016321_at
    • Gene symbols: KRT6B, DSC2, DSG3, FAM106B, KRT6A, KRT14, SPRR2D, RALA, SERPINB5, RHCG

The scores derived from these 10-genes are correlated to the original scores at the level of 0.99 for immune signature and 0.89 for the hypoxia signature.

The same model (with the same parameters) was used as Formula 27 for the reduced genesets to estimate the risk score. Table 60 lists number of samples, number of deaths, and the death rate in each prediction score bin.

TABLE 60 Average death rate versus prediction score. Number of Number of Score samples death Death Rate <0.4 15 7 0.47 0.4-0.6 51 32 0.63 0.6-0.8 50 44 0.88 >0.8 20 16 0.80

Using a threshold of 0.5, the odds ratio for overall survival is 3.7 (95% CI: 1.7-8.1), Fisher's Exact Test p-value=1.7×10.

Patients can be further divided into good (risk score<0.5), medium (score 0.5-0.75) and poor (score>0.75) prognosis groups. FIG. 57 shows the Kaplan-Meier curves for these 3 groups. The Chi-square on 2 degrees of freedom is 12.2 (P=2.2×10−3).

TABLE 57 Prognosis signature component 1 (anti- correlated with poor outcome) Probe Gene merck-NM_005356_at LCK merck-NM_006144_at GZMA merck-NM_014207_at CD5 merck-NM_005608_at PTPRCAP merck-NM_007181_at MAP4K1 merck-NM_002738_at PRKCB merck-Y00638_s_at PTPRC merck-BC014239_s_at PTPRC merck-NM_130446_at KLHL6 merck-NM_005546_at ITK CYFIP2 merck-NM_006257_at PRKCQ merck-NM_002104_at GZMK merck-NM_001504_at CXCR3 merck-NM_001001895_at UBASH3A merck-NM_002832_at PTPN7 merck-NM_018460_at ARHGAP15 merck-NM_001838_at CCR7 merck-NM_002209_at ITGAL merck-NM_006725_at CD6 merck-BC028068_s_at JAK3 INSL3 merck-NM_001079_at ZAP70 merck-NM_005541_at INPP5D merck-ENST00000318430_s_at TMC8 merck-NM_006564_at CXCR6 merck-NM_007237_s_at SP140 merck-NM_178129_at P2RY8 merck-NM_000647_s_at CCR2 merck-BU428565_s_at P2RY8 merck-NM_002351_s_at SH2D1A merck-NM_001040033_at CD53 merck-NM_005816_at CD96 merck-NM_198517_at TBC1D10C merck-NM_000733_at CD3E merck-NM_002163_at IRF8 merck-NM_000655_at SELL merck-NM_003037_at SLAMF1 merck-NM_003151_a_at STAT4 merck-NM_001007231_s_at ARHGAP25 merck-NM_018326_at GIMAP4 merck-NM_000377_at WAS merck-NM_001558_at IL10RA merck-NM_002985_at CCL5 merck-DT807100_at CD3D CD3G merck-NM_001465_at FYB merck-BP339517_a_at FYB merck-NM_030767_at AKNA merck-NM_005565_at LCP2 merck-NM_001040031_at CD37 merck-NM_002872_at RAC2 merck-NM_019604_at CRTAM merck-NM_005263_at GFI1 merck-NM_001037631_at CTLA4 ICOS merck-NM_016388_at TRAT1 merck-NM_014450_at SIT1 RMRP merck-NM_000732_at CD3D merck-NM_000073_at CD3G merck-NM_007360_at KLRK1 KLRC4-KLRK1 merck-NM_013351_at TBX21 merck-NM_032214_at SLA2 merck-NM_000639_at FASLG merck-NM_001242_at CD27 merck-ENST00000381961_at IL7R merck-NM_153206_s_at AMICA1 merck-NM_001025598_at ARHGAP30 USF1 merck-NM_001768_at CD8A merck-NM_003978_at PSTPIP1 merck-NM_014716_at ACAP1 merck-AK128740_s_at ILI6 merck-NM_006060_a_at IKZF1 merck-BC075820_at IKZF1 merck-NM_016293_at BIN2 merck-NM_012092_at ICOS merck-NM_005442_at EOMES LOC100996624 merck-NM_007074_at CORO1A merck-NM_000206_at IL2RG merck-NM_005041_at PRF1 merck-NM_024898_s_at DENND1C CRB3 merck-NM_173799_at TIGIT merck-NM_001767_at CD2 merck-NM_002348_at LY9 merck-X60502_s_at SPN QPRT merck-NM_153236_at GIMAP7 merck-NM_005601_at NKG7 merck-NM_032496_at ARHGAP9 merck-NM_004877_at GMFG merck-NM_021181_at SLAMF7 merck-NM_018384_at GIMAP5 GIMAP1-GIMAP5 merck-NM_181780_at BTLA merck-NM_001017373_at SAMD3 merck-NM_000734_at CD247 merck-NM_003650_at CST7 merck-NM_172101_at CD8B merck-NM_001803_at CD52 merck-NM_001778_at CD48 merck-NM_001025265_at CXorf65 merck-NM_198929_at PYHIN1 merck-ENST00000379833_at GVINP1 merck-NM_052931_at SLAMF6 merck-NM_001024667_s_at FCRL3 merck-NM_002258_at KLRB1 merck-NM_018556_s_at SIRPG merck-AK090431_s_at NLRC3 merck-NM_018990_at SASH3 XPNPEP2 merck-NM_175900_s_at C16orf54 QPRT merck-ENST00000316577_s_at TESPA1 merck-NM_024070_at PVRIG merck-AY190088_s_at merck-NM_001040067_s_at TRBC2 TRBV3-1 TRBV5-4 TRBV6-5 TRBV7-2 merck-NM_130848_s_at C5orf20 merck-ENST00000381153_at C11orf21 merck-ENST00000382913_s_at TRAC TRAJ17 TRAV20 TRDV2 merck-BC030533_s_at TRBC1 TRBV19 merck-ENST00000244032_a_at ZNF831 merck-ENST00000371030_at ZNF831 merck-ENST00000343625_s_at RASAL3 merck-AF143887_at merck-AK128436_at IKZF3 merck-AI281804_at GPR174 merck-AF086367_at merck-CR598049_at LINC00426 merck-BM700951_at KLRK1 KLRC4-KLRK1 merck-BX648371_at LINC00861 merck-BC070382_at merck2-AW798052_at AKNA merck2-BX640915_at TIGIT merck2-BM678246_at CD37 merck2-NM_025228_at TRAF3IP3 merck2-XM_033379_at WDFY4 merck2-AJ515553_at AMICA1 merck2-BP262340_at IL16 merck2-AK225623_at DENND1C CRB3 merck2-AL833681_at CD96 merck2-BF111803_at ARHGAP15 merck2-BX406128_at CD3G merck2-NM_153701_at merck2-BC020657_at GIMAP4 merck2-AY185344_at PYHIN1 merck2-DR159064_at EOMES LOC100996624 merck2-ENST00000390420_at TRBV3-1 TRBV5-4 TRBV6-5 TRBV7-2 merck2-ENST00000390420_s_at merck2-NM_001010923_at THEMIS merck2-ENST00000390409_at TRBC1 TRBV19 merck2-AX721088_at merck2-ENST00000390393_at TRBV19 merck2-AW341086_at merck2-AA278761_at merck2-AA278761_x_at merck2-ENST00000390394_s_at merck2-AA669142_at merck2-AW007991_at PTPRC merck2-BG743900_at PRKCB merck2-X06318_at PRKCB merck2-BI519527_at IKZF1 merck2-ENST00000390537_s_at merck2-AY292266_x_at merck2-NM_005816_a_at CD96 merck2-NM_198196_a_at CD96 merck2-NM_001114380_x_at ITGAL merck2-NM_007237_a_at SP140 merck2-NM_007237_at SP140 merck2-NM_052931_at SLAMF6 merck2-NM_001558_at IL10RA merck2-NM_007360_at KLRK1 KLRC4-KLRK1 merck2-NM_002209_x_at ITGAL merck2-NM_175900_at C16orf54 QPRT

TABLE 58 Prognosis signature component 2 (correlated with poor outcome) probe Gene merck-NM_002627_at PFKP PITRM1 merck-NM_000302_at PLOD1 merck-NM_001216_at CA9 RMRP merck-ENST00000377093_at KIF1B merck-BC004202_a_at CHEK1 merck-NM_030949_at PPP1R14C merck-CR593119_a_at CLIC4 merck-NM_001255_s_at CDC20 merck-BG679113_s_at KRT6A KRT6B KRT6C merck-NM_002421_at MMP1 merck-BQ217236_a_at SERPINB5 merck-NM_001793_at CDH3 merck-NM_001238_at CCNE1 merck-BU597348_s_at SYNCRIP merck-NM_006516_at SLC2A1 merck-BX648425_a_at DSC2 merck-X15014_a_at RALA merck-NM_018685_at ANLN merck-CR614206_a_at ERO1L merck-NM_001124_at ADM merck-NM_015440_at MTHFD1L merck-ENST00000367307_a_at MTHFD1L merck-NM_058179_at PSAT1 merck-NM_031415_s_at GSDMC merck-NM_005557_x_at KRT16 merck-NM_053016_at PALM2 PALM2-AKAP2 merck-CR602579_a_at CTPS1 merck-NM_001428_s_at ENO1 merck-ENST00000305850_at CENPN CMC2 merck-NM_005978_at S100A2 merck-NM_018643_at TREM1 merck-NM_006505_at PVR merck-NM_080655_s_at MSANTD3 merck-NM_001012507_at CENPW merck-ENST00000258005_a_at NHSL1 merck-AK129763_at LINC00673 merck-XM_927868_s_at PGK1 merck-XM_928117_x_at FAM106B merck-AL359337_at ADM merck-AA148856_s_at SYNCRIP merck2-AI989728_at SERPINB5 merck2-DQ892208_at CA9 RMRP merck2-AK022036_at WWTR1 merck2-AA677426_at merck2-AA677426_s_at merck2-BC004856_at NCS1 merck2-BG252150_at PFKP merck2-BC007633_at AGO2 merck2-BG400371_at merck2-DQ891441_at merck2-NM_017522_AS_at LRP8 merck2-AF039652_at RNASEH1 merck2-AV714642_at ANLN merck2-AB030656_at CORO1C merck2-NM_000291_at PGK1 merck2-NM_005554_at KRT6A merck2-BC002829_at S100A2 merck2-BU681245_at merck2-AK225899_a_at CTPS1 merck2-BC062635_a_at XPO5 merck2-AF257659_a_at CALU merck2-CA308717_at merck2-X56807_at DSC2 merck2-CR936650_at ANLN merck2-AY423725_a_at PGK1 merck2-BC103752_a_at PGK1

Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of skill in the art to which the disclosed invention belongs. Publications cited herein and the materials for which they are cited are specifically incorporated by reference.

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

1. A method for predicting prognosis of a patient with breast cancer, comprising:

(a) determining from a tumor biopsy sample from the subject gene expression intensities for each of the following categories of signature genes: (1) estrogen receptor (ER), (2) human epidermal growth factor receptor 2 (HER2), (3) at least 5 proliferation signature genes listed in Table 1, and (4) at least 5 immune signature genes listed in Table 2; and
(b) calculating a breast cancer risk score from the gene expression intensities;
wherein a high breast cancer risk score is an indication that the subject has a high risk for bone metastasis and death.

2. The method of claim 1, wherein the at least 5 proliferation signature genes are selected from the group consisting of TPX2, CENPA, KIF2C, CCNB2, BUB1, HJURP, CDCA5, CEP55, and SKA1

3. The method of claim 1, wherein the at least 5 immune signature genes are selected from the group consisting of CD3D, CD2, CD3E, ITK, TRBC1, TBC1D10C, ACAP1, CD247, SLAMF6, and IKZF1.

4. The method of claim 1, further comprising treating the subject with more aggressive treatment if the subject has a high breast cancer risk score.

5. A method for predicting prognosis of a patient with lung cancer, comprising: wherein a high lung cancer risk score is an indication that the subject has a high risk of death.

(a) determining from a tumor biopsy sample from the subject gene expression intensities for each of the following categories of signature genes:
(1) at least 5 immune signature genes listed in Table 4,
(2) at least 5 hypoxia signature genes listed in Table 5,
(3) at least 5 lung cancer prognosis signature genes listed in Table 7, and
(4) at least 5 proliferation signature genes listed in Table 8;
(b) determining the composite tumor stage; and
(c) calculating a lung cancer risk score from the gene expression intensities and composite tumor stage;

6. The method of claim 5, wherein the at least 5 immune signature genes are selected from the group consisting of CD2, ITGAL, IKZF1, CD3D, TRBC1, ACAP1, CD3E, TBC1D10C, CD247, and SLAMF6.

7. The method of claim 5, wherein the at least 5 hypoxia signature genes are selected from the group consisting of SLC2A1, S100A2, KRT16, KRT6A, CD109, GJB3, SFN, MICALL1, RNTL2, and COL7A1.

8. The method of claim 5, wherein the at least 5 lung cancer prognosis signature genes are selected from the group consisting of HLF, SCN7A, NR3C2, PCDP1, ABCA8, EMCN, IFT57, BDH2, MAMDC2, and ITGA8.

9. The method of claim 5, wherein the at least 5 proliferation signature genes are selected from the group consisting of TPX2, CENPA, KIF2C, CCNB2, CDCA5, HJURP, KIF4A, BIRC5, DLGAP5, and SKA1.

10. The method of claim 5 further comprising treating the subject with more aggressive treatment if the subject has a high lung cancer risk score.

11. A method for predicting prognosis of a patient with colon cancer, comprising:

(a) determining from a tumor biopsy sample from the subject gene expression intensities for each of the following categories of signature genes: (1) at least 5 immune signature genes listed in Table 12, (2) at least 5 hypoxia signature genes listed in Table 13, (3) at least 5 vimentin (VIM) correlated genes listed in Table 14, (4) at least 5 CDH1 correlated genes listed in Table 15, (5) at least 5 first prognosis signature genes listed in Table 16, and (6) at least 5 second prognosis signature genes listed in Table 17;
(b) determining the composite tumor stage; and
(c) calculating a colon cancer risk score from the gene expression intensities and composite tumor stage; wherein a high colon cancer risk score is an indication that the subject has a high risk of death.

12. The method of claim 11, wherein the at least 5 immune signature genes are selected from the group consisting of IKZF1, ITGAL, CD2, ITK, MAP4K1, CD3E, TBC1D10C, TRBC2, CD247, and CD3D.

13. The method of claim 11, wherein the at least 5 hypoxia signature genes are selected from the group consisting of SLC2A1, RALA, ERO1L, ANLN, S100A2, PHLDA2, CDC20, LAMC2, PLAUR, and SLC16A3.

14. The method of claim 11, wherein the at least 5 vimentin (VIM) correlated genes are selected from the group consisting of CCDC80, VIM, HEG1, CNRIP1, RAB31, EFEMP2, GNB4, MRAS, CMTM3, and TIMP2.

15. The method of claim 11, wherein the at least 5 CDH1 correlated genes are selected from the group consisting of ELF3, CLDN7, CLDN4, CDH1, RAB25, ESRP1, ESRP2, ERBB3, AP1M2, and EPCAM.

16. The method of claim 11, wherein the at least 5 first prognosis signature genes are selected from the group consisting of MZB1, OR6C4 IGKV3-11 IGKV3D-11 IGKV3D-20 RHNO1, TNFRSF17, IGKC IGKV1D-39 IGKV1-39, IGHA1 IGHG1 IGH, IGLC1, IGKC IGKV1-16 IGKV1D-16, IGLV6-57, IGLV1-40 IGLV5-39, and IGJ.

17. The method of claim 11, wherein the at least 5 second prognosis signature genes are selected from the group consisting of SPP1, CDH2, ITGB1, SERPINE1, PLOD2, COL4A1, NTM, MPRIP, PLIN2, and TIMP1.

18. The method of claim 11, further comprising treating the subject with more aggressive treatment if the subject has a high colon cancer risk score.

19. A method for predicting prognosis of a patient with kidney cancer, comprising:

(a) determining from a tumor biopsy sample from the subject gene expression intensities for each of the following categories of signature genes: (1) at least 5 first prognosis signature genes listed in Table 22, and (2) at least 5 second prognosis signature genes listed in Table 23; and
(b) calculating a kidney cancer risk score from the gene expression intensities; wherein a high kidney cancer risk score is an indication that the subject has a high risk of death.

20. The method of claim 19, wherein the at least 5 first prognosis signature genes are selected from the group consisting of CRY2, NR3C2, HLF, EMX2OS, FAM221B, BDH2, BCL2, ACADL, NDRG2, and NPR3.

21. The method of claim 19, wherein the at least 5 second prognosis signature genes are selected from the group consisting of TPX2, CCNB2, AURKB, HJURP, CENPA, CENPF, SKA1, CEP55, PTTG1, and FOXM1

22. The method of claim 19, further comprising treating the subject with more aggressive treatment if the subject has a high kidney cancer risk score.

23-70. (canceled)

Patent History
Publication number: 20170298443
Type: Application
Filed: Sep 24, 2015
Publication Date: Oct 19, 2017
Inventor: Hongyue A. Dai (Chesnut Hill, MA)
Application Number: 15/514,147
Classifications
International Classification: C12Q 1/68 (20060101);